1
|
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
|
2
|
Ambati S, Choudhury QJ, Peter JA, Moremen KW, Chapla DG, Lewis ZA, Lin X, Meagher RB. Siglec-targeted liposomes to identify sialoglycans present on fungal pathogens. Antimicrob Agents Chemother 2025; 69:e0172024. [PMID: 40084878 PMCID: PMC11963605 DOI: 10.1128/aac.01720-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/16/2025] [Indexed: 03/16/2025] Open
Abstract
The sialic acid Ig-like lectins Siglec-3 and Siglec-15 are pathogen receptors that bind sialic acid-modified glycoproteins, best characterized in metastatic cancers. Because fungi produce sialoglycans and sialo-glycoproteins, we wondered if Siglecs had the potential for targeted delivery of antifungal drugs. We purified the extracellular V-region Ig-like C2 ligand-binding domains and stalk regions of SIG3 and SIG15. We floated the two polypeptides on the surface of liposomes loaded with amphotericin B (AmB) and labeled with rhodamine B to prepare SIG3-Ls and SIG15-Ls. Using these two reagents, we explored the sialoglycans of two evolutionarily distant and deadly human fungal pathogens, the Mucormycete Rhizopus delemar and the Ascomycete Aspergillus fumigatus. We found that SIG3-Ls and SIG15-Ls localized in a continuous layer over the cell wall surface of germ tubes and hyphae of both fungal species and to the conidia of A. fumigatus. Binding was Neu5Ac-specific and appeared confined to N-linked sialoglycans on fungal proteins. SIG3 and SIG15 proteins bound to diverse sialo-glycoproteins extracted from the hyphae of both species. SIG3-Ls and SIG15-Ls delivering sub-micromolar concentrations of AmB were moderately more effective at inhibiting and/or killing both species relative to control liposomes. We discuss the roles that sialo-glycoproteins may play in fungal pathogens.
Collapse
Affiliation(s)
- Suresh Ambati
- Department of Genetics, University of Georgia, Athens, Georgia, USA
| | | | - Jesse Ann Peter
- Department of Genetics, University of Georgia, Athens, Georgia, USA
| | - Kelley W. Moremen
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Digantkumar Gopaldas Chapla
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Zachary A. Lewis
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | - Xiaorong Lin
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
| | | |
Collapse
|
3
|
Wang J, Li Z, He Y, Chu Y. Targeting Siglec-15 mediates mitochondrial retrograde regulation of cervical cancer development. Tissue Cell 2025; 93:102713. [PMID: 39756115 DOI: 10.1016/j.tice.2024.102713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/07/2025]
Abstract
Cervical cancer (CCA) is the predominant cause of fatalities from gynecologic malignancies, with metastasis responsible for 80 % of cancer-related mortalities. This study preliminarily examined the involvement of Sialic Acid Binding Ig Like Lectin 15 (Siglec-15) in the development of CCA and its probable mechanisms. We assessed the capacity of Siglec-15 to modulate CCA progression by establishing knockdown and overexpression Siglec-15 cell lines, supplemented with animal models, using both in vivo and in vitro dual investigations. Our findings indicate that Siglec-15 is significantly expressed in CCA cell lines and is intimately associated with the proliferation, migration, and invasion capabilities of CCA cells, as well as mitochondrial ROS homeostasis. The suppression of Siglec-15 expression markedly reduced tumor growth in mice, potentially due to Siglec-15's role in regulating the Mitogen-Activated Protein Kinase (MAPK) signaling pathway, which mediates the retrograde regulation of mitochondrial ROS homeostasis. Siglec-15 may emerge as a novel therapeutic target and prognostic marker for patients with CCA.
Collapse
Affiliation(s)
- Jing Wang
- Department of Gynecology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong 264000, China
| | - Zenghui Li
- Department of Gynecology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, China
| | - Yifan He
- Department of Radiology, Yantaishan Hospital, Yantai, Shandong 264000, China
| | - Yongli Chu
- Department of Gynecology, Yantai Yuhuangding Hospital, Shandong University, Yantai, Shandong 264000, China.
| |
Collapse
|
4
|
Fan Y, Sun L, He J, Chen Y, Ma H, Ding H. Siglec15 in blood system diseases: from bench to bedside. Front Immunol 2024; 15:1490505. [PMID: 39697338 PMCID: PMC11652361 DOI: 10.3389/fimmu.2024.1490505] [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: 09/03/2024] [Accepted: 11/13/2024] [Indexed: 12/20/2024] Open
Abstract
Inhibiting the PD-1/PD-L1 pathway using immunomodulators has demonstrated promising outcomes in clinics. Immunomodulators can effectively target immune checkpoints with a strong preference for the tumor microenvironment (TME). Besides, immunomodulators specifically target the recently discovered inhibitory immune checkpoint, sialic acid-binding immunoglobulin-like lectin (Siglec-15). Distinctive in its molecular composition, Siglec-15 has a unique molecular composition and been shown to be highly prevalent in numerous solid tumor tissues and tumor-associated macrophages (TAMs) in human subjects. Notably, Siglec-15 is up-regulated across various cancer types. As a result, Siglec-15 has attracted significant attention due to its exclusive nature concerning PD-L1 expression, suggesting its role in immune evasion in patients lacking PD-L1. Siglec-15 predominantly appears in certain populations and can promote tumor development by repressing T lymphocyte activation and proliferation, thereby facilitating tumor cell immune escape. Furthermore, Siglec-15 is implicated in osteoclast differentiation and bone remodeling, indicating that it is a promising target for next-generation cancer immunotherapies. Additionally, Siglec-15 can modulate immune responses to microbial infections. The current treatment strategies for hematological conditions predominantly include conventional intensive chemotherapy and transplantation methods. However, emerging immunotherapeutic approaches are increasingly recognized for their overall effectiveness, indicating that specific molecular targets should be identified. The expression of Siglec-15 within tumor cells may indicate a novel pathway for treating hematological malignancies. In this study, the biological attributes, expression patterns, and pathogenic mechanisms of Siglec-15 across various diseases were reviewed. The role of Siglec-15 in the pathogenesis and laboratory diagnosis of hematological disorders was also evaluated.
Collapse
Affiliation(s)
- Yujia Fan
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Liangliang Sun
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Juan He
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Yuetong Chen
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| | - Hongli Ma
- Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, Inner Mongolia, China
| | - Haitao Ding
- Clinical Laboratory Medicine Centre, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
5
|
Liu C, Zhong Y, Huang H, Lan S, Li J, Huang D, Zhang W. Killing two birds with one stone: Siglec-15 targeting integrated bioactive glasses hydrogel for treatment of breast cancer bone metastasis. Mater Today Bio 2024; 29:101362. [PMID: 39687802 PMCID: PMC11647236 DOI: 10.1016/j.mtbio.2024.101362] [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: 08/11/2024] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Bone metastasis is a fatal consequence of breast cancer that occurs when patients fail to respond to conventional therapies and mainly result from a vicious cycle involving dysregulated bone homeostasis and uncontrolled tumor growth. Recent research has underscored the significance of Siglec-15, a membrane protein implicated in immunosuppression and osteoclast generation. Targeting Siglec-15 may disrupt the "vicious cycle" that causes bone metastases in patients with breast cancer. Herein, we explored the efficacy of targeting Siglec-15 in conjunction with photothermal chemotherapy to impede the progression of bone metastatic during breast cancer and repair tumor-induced osteolysis. First, we formulated an injectable photothermal bioactive glass (BG)-based hydrogel for the local delivery of Siglec-15 shRNA and doxorubicin. The results demonstrated that the hydrogel could kill tumor cells directly through photothermal chemotherapy, provoke intense immune responses and improve the local immunosuppressive microenvironment, which could effectively prevent tumor metastasis and recurrence in a murine model. The combined effect of BGs and Siglec15 shRNA can normalize dysregulated bone homeostasis at the bone metastasis site and significantly reduced bone destruction. Overall, the use of Siglec-15-targeting integrated BG hydrogels may provide a promising therapeutic strategy for treating bone metastasis caused by breast cancer.
Collapse
Affiliation(s)
- Chengkuan Liu
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Yangui Zhong
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Haibo Huang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Siyuan Lan
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Jing Li
- Second People's Hospital of Shenzhen, Shenzhen, Guangdong, PR China
| | - Deqiu Huang
- School of Medical Information Engineering, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Intelligent Chinese Medicine Research Institute, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| | - Wen Zhang
- Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, PR China
| |
Collapse
|
6
|
Lu J, Zhang Y, Wen H, Li J, Chen C, Xiao L. Siglec-15 as a potential molecule involved in osteoclast differentiation and bone metabolism. Heliyon 2024; 10:e38537. [PMID: 39524871 PMCID: PMC11550040 DOI: 10.1016/j.heliyon.2024.e38537] [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: 09/21/2023] [Revised: 08/29/2024] [Accepted: 09/25/2024] [Indexed: 11/16/2024] Open
Abstract
Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) is a well-conserved type I transmembrane protein of the Siglecs family, distributed in macrophages and dendritic cells in the human spleen and lymph nodes. As an immune receptor, Siglec-15 is expressed in almost all branches of the spinal cord. Siglec-15 participates in the metabolism of the skeleton by regulating osteoclast activity and differentiation and has an influential role in dynamic bone remodelling. The binding of DNAX activation protein of 12 kDa (DAP12), which contains the immunoreceptor tyrosine-based activation motif (ITAM) activation domain, to the Siglec-15 receptor provides a positive stimulatory signal for osteoclast growth, with the involvement of the receptor activator of nuclear factor-κB (RANK)/RANK ligand (RANKL) signalling. Recently, Siglec-15 antibodies have been shown to effectively prevent bone resorption in mouse models of osteoporosis and accelerate fracture healing to some extent. Therefore, exploring the molecular characteristics and functions of Siglec-15 may lead to new therapeutic strategies for common clinical skeletal diseases.
Collapse
Affiliation(s)
- Jiaqi Lu
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Yinyin Zhang
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Huiyu Wen
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Junlin Li
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Chen Chen
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| | - Liwei Xiao
- Department of Orthodontics, Medical Center of Stomatology, The Second Xiangya Hospital, Central South University, Furong District, Changsha, Hunan, 410011, China
| |
Collapse
|
7
|
Shen L, Schaefer AM, Tiruthani K, Wolf W, Lai SK. Siglec15/TGF-β bispecific antibody mediates synergistic anti-tumor response against 4T1 triple negative breast cancer in mice. Bioeng Transl Med 2024; 9:e10651. [PMID: 39553435 PMCID: PMC11561775 DOI: 10.1002/btm2.10651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 11/19/2024] Open
Abstract
An ideal tumor-specific immunomodulatory therapy should both preferentially target the tumor, while simultaneously reduce the immunosuppressive environment within the tumor. This guiding principle led us to explore engineering Siglec-15 (S15) targeted bispecific antibody (bsAb) to enhance therapy against triple negative breast cancer (TNBC). S15 appears to be exclusively expressed on macrophages and diverse tumor cells, including human and mouse 4T1 TNBC. TGF-β is a growth hormone frequently associated with increased tumor invasiveness, including in TNBC. Here, to overcome the immune-suppressive environment within TNBC tumors to enable more effective cancer therapy, we engineered a bispecific antibody (bsAb) targeting both Siglec15 and TGF-β. In mice engrafted with orthotopic 4T1 tumors, S15/TGF-β bsAb treatment was highly effective in suppressing tumor growth, not only compared to control monoclonal antibody (mAb) but also markedly more effective than mAbs against S15 alone, against TGF-β alone, as well as a cocktail of both anti-S15 and anti-TGF-β mAbs. We did not detect liver and lung metastasis in mice treated with S15/TGF-β bsAb, unlike all other treatment groups at the end of the study. The enhanced anti-tumor response observed with S15/TGF-β bsAb correlated with a less immunosuppressive environment in the tumor. These results underscore S15-targeted bsAb as a promising therapeutic strategy for TNBC, and possibly other S15 positive solid tumors.
Collapse
Affiliation(s)
- Limei Shen
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Alison M. Schaefer
- Department of Biomedical EngineeringUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Karthik Tiruthani
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Whitney Wolf
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
| | - Samuel K. Lai
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of Pharmacy, University of North CarolinaChapel HillNorth CarolinaUSA
- Department of Biomedical EngineeringUniversity of North CarolinaChapel HillNorth CarolinaUSA
- Department of Immunology and MicrobiologyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| |
Collapse
|
8
|
Zhang Y, Sun H, Huang F, Chen Y, Ding X, Zhou C, Wu Y, Zhang Q, Ma X, Wang J, Yue R, Shen L, Sun X, Ye Z. The chromatin remodeling factor Arid1a cooperates with Jun/Fos to promote osteoclastogenesis by epigenetically upregulating Siglec15 expression. J Bone Miner Res 2024; 39:775-790. [PMID: 38477755 DOI: 10.1093/jbmr/zjae042] [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: 06/23/2023] [Revised: 02/09/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Osteoporosis is characterized by an imbalance between osteoclast-mediated bone resorption and osteoblast-related bone formation, particularly increased osteoclastogenesis. However, the mechanisms by which epigenetic factors regulate osteoclast precursor differentiation during osteoclastogenesis remain poorly understood. Here, we show that the specific knockout of the chromatin remodeling factor Arid1a in bone marrow-derived macrophages (BMDMs) results in increased bone mass. The loss of Arid1a in BMDM inhibits cell-cell fusion and maturation of osteoclast precursors, thereby suppressing osteoclast differentiation. Mechanistically, Arid1a increases the chromatin access in the gene promoter region of sialic acid-binding Ig-like lectin 15 (Siglec15) by transcription factor Jun/Fos, which results in the upregulation of Siglec15 and promotion of osteoclast differentiation. However, the loss of Arid1a reprograms the chromatin structure to restrict Siglec15 expression in osteoclast precursors, thereby inhibiting BMDM differentiation into mature osteoclasts. Deleting Arid1a after ovariectomy (a model for postmenopausal bone loss) alleviated bone loss and maintained bone mass. In summary, epigenetic reprogramming mediated by Arid1a loss suppresses osteoclast differentiation and may serve as a promising therapeutic strategy for treating bone loss diseases.
Collapse
Affiliation(s)
- Yongxing Zhang
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
| | - Hangxiang Sun
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
| | - Fei Huang
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Yang Chen
- Department of Ultrasound, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, PR China
| | - Xiying Ding
- Department of Ultrasound, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, PR China
| | - Chenhe Zhou
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
| | - Yan Wu
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
| | - Qing Zhang
- Research Institute of Artificial Intelligence, Zhejiang Lab, Hangzhou, Zhejiang 311121, PR China
| | - Xiao Ma
- Research Institute of Artificial Intelligence, Zhejiang Lab, Hangzhou, Zhejiang 311121, PR China
| | - Jun Wang
- Research Institute of Artificial Intelligence, Zhejiang Lab, Hangzhou, Zhejiang 311121, PR China
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai 200092, PR China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, PR China
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
- Hangzhou Innovation Center, Zhejiang University, Hangzhou, Zhejiang 310058, PR China
| | - Xuxu Sun
- Department of Biochemistry and Molecular Cell Biology, State Key Laboratory of Systems Medicine for Cancer, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Zhaoming Ye
- Department of Orthopedics, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, PR China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang 310009, PR China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang 310009, PR China
| |
Collapse
|
9
|
Zhao K, Wu C, Li X, Niu M, Wu D, Cui X, Zhao H. From mechanism to therapy: the journey of CD24 in cancer. Front Immunol 2024; 15:1401528. [PMID: 38881902 PMCID: PMC11176514 DOI: 10.3389/fimmu.2024.1401528] [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: 03/15/2024] [Accepted: 04/25/2024] [Indexed: 06/18/2024] Open
Abstract
CD24 is a glycosylphosphatidylinositol-anchored protein that is expressed in a wide range of tissues and cell types. It is involved in a variety of physiological and pathological processes, including cell adhesion, migration, differentiation, and apoptosis. Additionally, CD24 has been studied extensively in the context of cancer, where it has been found to play a role in tumor growth, invasion, and metastasis. In recent years, there has been growing interest in CD24 as a potential therapeutic target for cancer treatment. This review summarizes the current knowledge of CD24, including its structure, function, and its role in cancer. Finally, we provide insights into potential clinical application of CD24 and discuss possible approaches for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Caifeng Wu
- Department of Hand and Foot, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiangjun Li
- Department of Breast Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Mengchao Niu
- Department of Operation Room, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Wu
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaofeng Cui
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
10
|
Dedola S, Ahmadipour S, de Andrade P, Baker AN, Boshra AN, Chessa S, Gibson MI, Hernando PJ, Ivanova IM, Lloyd JE, Marín MJ, Munro-Clark AJ, Pergolizzi G, Richards SJ, Ttofi I, Wagstaff BA, Field RA. Sialic acids in infection and their potential use in detection and protection against pathogens. RSC Chem Biol 2024; 5:167-188. [PMID: 38456038 PMCID: PMC10915975 DOI: 10.1039/d3cb00155e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/12/2023] [Indexed: 03/09/2024] Open
Abstract
In structural terms, the sialic acids are a large family of nine carbon sugars based around an alpha-keto acid core. They are widely spread in nature, where they are often found to be involved in molecular recognition processes, including in development, immunology, health and disease. The prominence of sialic acids in infection is a result of their exposure at the non-reducing terminus of glycans in diverse glycolipids and glycoproteins. Herein, we survey representative aspects of sialic acid structure, recognition and exploitation in relation to infectious diseases, their diagnosis and prevention or treatment. Examples covered span influenza virus and Covid-19, Leishmania and Trypanosoma, algal viruses, Campylobacter, Streptococci and Helicobacter, and commensal Ruminococci.
Collapse
Affiliation(s)
- Simone Dedola
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Sanaz Ahmadipour
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Peterson de Andrade
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Alexander N Baker
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Andrew N Boshra
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University Assiut 71526 Egypt
| | - Simona Chessa
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Matthew I Gibson
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
- Division of Biomedical Sciences, Warwick Medical School Coventry CV4 7AL UK
| | - Pedro J Hernando
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Irina M Ivanova
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Jessica E Lloyd
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - María J Marín
- School of Chemistry, University of East Anglia, Norwich Research Park Norwich NR4 7TJ UK
| | - Alexandra J Munro-Clark
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | | | - Sarah-Jane Richards
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Department of Chemistry, University of Warwick Gibbet Hill Road Coventry CV4 7AL UK
| | - Iakovia Ttofi
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| | - Ben A Wagstaff
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| | - Robert A Field
- Department of Chemistry and Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
- Iceni Glycoscience Ltd, Norwich Research Park Norwich NR4 7TJ UK
| |
Collapse
|
11
|
Wang H, Shi P, Shi X, Lv Y, Xie H, Zhao H. Surprising magic of CD24 beyond cancer. Front Immunol 2024; 14:1334922. [PMID: 38313430 PMCID: PMC10834733 DOI: 10.3389/fimmu.2023.1334922] [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: 11/08/2023] [Accepted: 12/28/2023] [Indexed: 02/06/2024] Open
Abstract
CD24 has emerged as a molecule of significant interest beyond the oncological arena. Recent studies have unveiled its surprising and diverse roles in various biological processes and diseases. This review encapsulates the expanding spectrum of CD24 functions, delving into its involvement in immune regulation, cancer immune microenvironment, and its potential as a therapeutic target in autoimmune diseases and beyond. The 'magic' of CD24, once solely attributed to cancer, now inspires a new paradigm in understanding its multifunctionality in human health and disease, offering exciting prospects for medical advancements.
Collapse
Affiliation(s)
- He Wang
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Shi
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinyu Shi
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yaqing Lv
- Department of Outpatient, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongwei Xie
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hai Zhao
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Moreira RS, da Silva MM, de Melo Vasconcelos CF, da Silva TD, Cordeiro GG, Mattos-Jr LAR, da Rocha Pitta MG, de Melo Rêgo MJB, Pereira MC. Siglec 15 as a biomarker or a druggable molecule for non-small cell lung cancer. J Cancer Res Clin Oncol 2023; 149:17651-17661. [PMID: 37843557 DOI: 10.1007/s00432-023-05437-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023]
Abstract
Lung cancer has been the main cause of cancer mortality worldwide. Furthermore, lung cancer rates of new cases per year evidenced a large incidence of this neoplasm in both men and women. Because there is no biomarker for early detection, it is frequently detected late, at an advanced state. The introduction of multiple lines of tyrosine kinase inhibitors in patients with EGFR, ALK, ROS1, and NTRK mutations has modified the therapy of lung cancer. Immunotherapy advances have resulted in substantial improvements in overall survival and disease-free survival, making immune checkpoint inhibitors (ICIs) a potential option for lung cancer treatment. Current PD-1/PD-L1/CTLA-4 immunotherapies have resulted in important response and survival rates. However, existing medicines only function in around 20% of unselected, advanced NSCLC patients, and primary and acquired resistance remain unsolved obstacles. Therefore, precise predictive indicators must be identified to choose the best patients for ICI treatment. Thus, Sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) stands out as a potential tumor biomarker, with distinctive expression in normal tissues, in tumor immune involvement, and a high structural similarity to PD-L1. Understanding the tumor immune response and the search for new therapeutic targets leads to the improvement of therapeutic pathways directed at the tumor microenvironment. The present review aims to analyze Siglec-15 potential as a diagnostic, prognostic, and response biomarker in lung cancer, considering its results evidenced in the current literature.
Collapse
Affiliation(s)
- Rodrigo Santiago Moreira
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | - Marillya Morais da Silva
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Thiago Douberin da Silva
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Luiz Alberto Reis Mattos-Jr
- Department of Clinic Medicine, Federal University of Pernambuco, Av. Prof. Moraes Rego, 1235, Recife, PE, Brazil
| | - Maira Galdino da Rocha Pitta
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil
| | | | - Michelly Cristiny Pereira
- Suely-Galdino Therapeutic Innovation Research Center (NUPIT-SG), Federal University of Pernambuco, Recife, Pernambuco, Brazil.
| |
Collapse
|
13
|
Yang S, He Z, Wu T, Wang S, Dai H. Glycobiology in osteoclast differentiation and function. Bone Res 2023; 11:55. [PMID: 37884496 PMCID: PMC10603120 DOI: 10.1038/s41413-023-00293-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 08/20/2023] [Accepted: 09/07/2023] [Indexed: 10/28/2023] Open
Abstract
Glycans, either alone or in complex with glycan-binding proteins, are essential structures that can regulate cell biology by mediating protein stability or receptor dimerization under physiological and pathological conditions. Certain glycans are ligands for lectins, which are carbohydrate-specific receptors. Bone is a complex tissue that provides mechanical support for muscles and joints, and the regulation of bone mass in mammals is governed by complex interplay between bone-forming cells, called osteoblasts, and bone-resorbing cells, called osteoclasts. Bone erosion occurs when bone resorption notably exceeds bone formation. Osteoclasts may be activated during cancer, leading to a range of symptoms, including bone pain, fracture, and spinal cord compression. Our understanding of the role of protein glycosylation in cells and tissues involved in osteoclastogenesis suggests that glycosylation-based treatments can be used in the management of diseases. The aims of this review are to clarify the process of bone resorption and investigate the signaling pathways mediated by glycosylation and their roles in osteoclast biology. Moreover, we aim to outline how the lessons learned about these approaches are paving the way for future glycobiology-focused therapeutics.
Collapse
Affiliation(s)
- Shufa Yang
- Prenatal Diagnostic Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100026, China
| | - Ziyi He
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China
| | - Tuo Wu
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China
| | - Shunlei Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100191, China.
| |
Collapse
|
14
|
Wu Y, Ai H, Xi Y, Tan J, Qu Y, Xu J, Luo F, Dou C. Osteoclast-derived apoptotic bodies inhibit naive CD8 + T cell activation via Siglec15, promoting breast cancer secondary metastasis. Cell Rep Med 2023; 4:101165. [PMID: 37607544 PMCID: PMC10518580 DOI: 10.1016/j.xcrm.2023.101165] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 04/18/2023] [Accepted: 07/31/2023] [Indexed: 08/24/2023]
Abstract
The bone microenvironment promotes cancer cell proliferation and dissemination. During periodic bone remodeling, osteoclasts undergo apoptosis, producing large numbers of apoptotic bodies (ABs). However, the biological role of osteoclast-derived ABs, which are residents of the bone-tumor niche, remains largely unknown. Here, we discover that AB-null MRL/lpr mice show resistance to breast cancer cell implantation, with more CD8+ T cell infiltrations and a higher survival rate. We uncover that the membranous Siglec15 on osteoclast-derived ABs binds with sialylated Toll-like receptor 2 (TLR2) and blocks downstream co-stimulatory signaling, leading to the inhibition of naive CD8+ T cell activation. In addition, our study shows that treatment with Siglec15 neutralizing antibodies significantly reduces the incidence of secondary metastases and improves the survival rate of mice with advanced breast cancer bone metastasis. Our findings reveal the immunosuppressive function of osteoclast-derived ABs in the bone-tumor niche and demonstrate the potential of Siglec15 as a common target for anti-resorption and immunotherapy.
Collapse
Affiliation(s)
- Yutong Wu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongbo Ai
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yuhang Xi
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jiulin Tan
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Ying Qu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianzhong Xu
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Fei Luo
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Ce Dou
- Department of Orthopedics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| |
Collapse
|
15
|
Ye Z, Wang Y, Xiang B, Wang H, Tao H, Zhang C, Zhang S, Sun D, Luo F, Song L. Roles of the Siglec family in bone and bone homeostasis. Biomed Pharmacother 2023; 165:115064. [PMID: 37413904 DOI: 10.1016/j.biopha.2023.115064] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/23/2023] [Accepted: 06/23/2023] [Indexed: 07/08/2023] Open
Abstract
Tremendous progress has been seen in the study of the role of sialic acid binding im-munoglobulin type lectins (Siglecs) in osteoimmunology in the past two decades. Interest in Siglecs as immune checkpoints has grown from the recognition that Siglecs have relevance to human disease. Siglecs play important roles in inflammation and cancer, and play key roles in immune cell signaling. By recognizing common sialic acid containing glycans on glycoproteins and glycolipids as regulatory receptors for immune cell signals, Siglecs are expressed on most immune cells and play important roles in normal homeostasis and self-tolerance. In this review, we describe the role that the siglec family plays in bone and bone homeostasis, including the regulation of osteoclast differentiation as well as recent advances in inflammation, cancer and osteoporosis. Particular emphasis is placed on the relevant functions of Siglecs in self-tolerance and as pattern recognition receptors in immune responses, thereby potentially providing emerging strategies for the treatment of bone related diseases.
Collapse
Affiliation(s)
- Zi Ye
- The Fourth Corps of Students of the Basic Medical College, Army Medical University, Chongqing 400037, China
| | - Yetong Wang
- The Fourth Corps of Students of the Basic Medical College, Army Medical University, Chongqing 400037, China
| | - Binqing Xiang
- Department of Surgical Anesthesia, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Heng Wang
- Army Border Defense 331st Brigade, Dandong 118000, China
| | - Haiyan Tao
- Health Management Center, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Chengmin Zhang
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Shuai Zhang
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China
| | - Dong Sun
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China.
| | - Fei Luo
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China.
| | - Lei Song
- Department of Orthopaedics, First Affiliated Hospital, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
16
|
Mutka M, Joensuu K, Heiskala M, Eray M, Heikkilä P. Core needle biopsies alter the amounts of CCR5, Siglec-15, and PD-L1 positivities in breast carcinoma. Virchows Arch 2023; 483:215-224. [PMID: 37222841 PMCID: PMC10412655 DOI: 10.1007/s00428-023-03563-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/25/2023]
Abstract
Core needle biopsies (CNB) are widely used to diagnose breast cancer, but the procedure is invasive and thus, it changes the tumor microenvironment. The purpose of this study is to see how the expression of three potentially anti-inflammatory molecules, namely, programmed death-ligand 1 (PD-L1), sialic acid-binding immunoglobulin-like lectin-15 (Siglec-15), and C-C chemokine receptor-5 (CCR-5), are expressed in CNB and surgical resection specimens (SRS). To do this, we compared the amounts of tumor-infiltrating lymphocytes and the levels of CCR5, Siglec-15, and PD-L1 in tumor cells and inflammatory cells as assessed by immunohistochemistry in CNB and the corresponding SRS of 22 invasive breast carcinomas of no special type and 22 invasive lobular carcinomas. The Siglec-15 H-score was higher in tumor cells in the SRS than in the CNB groups. There was no change in tumor cells CCR5 or PD-L1 between CNB and SRS. The positive inflammatory cell numbers for all markers rose between CNB and SRS, as did the amount of Tils. Furthermore, higher grade tumors and tumors with a high proliferation rate had more inflammatory cells that were positive for the markers and also more PD-L1+ tumor cells. Although changes in inflammatory cells can partly be attributed to the larger sample size of operation specimens, the differences also mirror a true change in the tumor microenvironment. The changes in inflammatory cells could be partly due to the need to restrict excess inflammation at the site of the biopsy.
Collapse
Affiliation(s)
- Minna Mutka
- Department of Pathology, HUSLAB, Helsinki University Hospital and University of Helsinki, FIN-00290, Helsinki, Finland.
| | | | | | - Mine Eray
- Department of Pathology, HUSLAB, Helsinki University Hospital and University of Helsinki, FIN-00290, Helsinki, Finland
| | - Päivi Heikkilä
- Department of Pathology, HUSLAB, Helsinki University Hospital and University of Helsinki, FIN-00290, Helsinki, Finland
| |
Collapse
|
17
|
Ding H, Yao B, Ci L, Feng J, Ouyang P, Chen G, Hui X, Zhou D. Enhancing the Anti-tumor Potency of a Novel Siglec-15 Antibody by Engineering its Fc-mediated Effector Functions. J Immunother 2023; 46:161-169. [PMID: 37103472 PMCID: PMC10168116 DOI: 10.1097/cji.0000000000000465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/22/2023] [Indexed: 04/28/2023]
Abstract
Siglec-15, an inhibitory immune checkpoint, is an emerging target in cancer immunotherapy. Blocking the function of Siglec-15 is an excellent strategy for cancer treatment and antibody blockade has been used to target Siglec-15. However, whether Fc-mediated effector functions contribute to the therapeutic effect of antibodies remains unclear. Herein, we generated a monoclonal antibody, 1-15D1, which had a high binding affinity with Siglec-15 and strongly activated T-cell immune response in vitro. Subsequently, the Fc-mediated effector functions of 1-15D1 were explored in a Siglec-15 humanized mouse model, and further improvement in antitumor efficacy was observed in the mouse IgG2a isotype group. Thus, we demonstrate that the antitumor effects of 1-15D1 were mediated via multiple factors. In addition to the T-cell immune response, 2 novel mechanisms were explored, including the internalization of the cell surface Siglec-15 and Fc-mediated effector functions. In conclusion, our studies not only provide a potential agent for the improvement of cancer immunotherapy but also suggest that a specific role of Fc-mediated immune regulation may improve the therapeutic potency of Siglec-15 monoclonal antibody.
Collapse
Affiliation(s)
- Huandi Ding
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing
- CSPC Megalith Biopharmaceutical Co. Ltd, Shijiazhuang
| | - Bing Yao
- CSPC Megalith Biopharmaceutical Co. Ltd, Shijiazhuang
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing
| | - Lei Ci
- Shanghai Engineering Research Center for Model Organisms, SMOC, Shanghai, China
| | - Jing Feng
- CSPC Megalith Biopharmaceutical Co. Ltd, Shijiazhuang
| | - Pingkai Ouyang
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing
| | - Guoguang Chen
- School of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing
| | - Xiwu Hui
- CSPC Megalith Biopharmaceutical Co. Ltd, Shijiazhuang
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing
| |
Collapse
|
18
|
Schmidt EN, Lamprinaki D, McCord KA, Joe M, Sojitra M, Waldow A, Nguyen J, Monyror J, Kitova EN, Mozaneh F, Guo XY, Jung J, Enterina JR, Daskhan GC, Han L, Krysler AR, Cromwell CR, Hubbard BP, West LJ, Kulka M, Sipione S, Klassen JS, Derda R, Lowary TL, Mahal LK, Riddell MR, Macauley MS. Siglec-6 mediates the uptake of extracellular vesicles through a noncanonical glycolipid binding pocket. Nat Commun 2023; 14:2327. [PMID: 37087495 PMCID: PMC10122656 DOI: 10.1038/s41467-023-38030-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 04/12/2023] [Indexed: 04/24/2023] Open
Abstract
Immunomodulatory Siglecs are controlled by their glycoprotein and glycolipid ligands. Siglec-glycolipid interactions are often studied outside the context of a lipid bilayer, missing the complex behaviors of glycolipids in a membrane. Through optimizing a liposomal formulation to dissect Siglec-glycolipid interactions, it is shown that Siglec-6 can recognize glycolipids independent of its canonical binding pocket, suggesting that Siglec-6 possesses a secondary binding pocket tailored for recognizing glycolipids in a bilayer. A panel of synthetic neoglycolipids is used to probe the specificity of this glycolipid binding pocket on Siglec-6, leading to the development of a neoglycolipid with higher avidity for Siglec-6 compared to natural glycolipids. This neoglycolipid facilitates the delivery of liposomes to Siglec-6 on human mast cells, memory B-cells and placental syncytiotrophoblasts. A physiological relevance for glycolipid recognition by Siglec-6 is revealed for the binding and internalization of extracellular vesicles. These results demonstrate a unique and physiologically relevant ability of Siglec-6 to recognize glycolipids in a membrane.
Collapse
Affiliation(s)
- Edward N Schmidt
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | | | - Kelli A McCord
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Maju Joe
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Mirat Sojitra
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ayk Waldow
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jasmine Nguyen
- Department of Obstetrics & Gynaecology and Physiology University of Alberta, Edmonton, AB, Canada
| | - John Monyror
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Elena N Kitova
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Fahima Mozaneh
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Xue Yan Guo
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jaesoo Jung
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Jhon R Enterina
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Gour C Daskhan
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ling Han
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Amanda R Krysler
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
| | | | - Basil P Hubbard
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Lori J West
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Marianne Kulka
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
- National Research Council, Edmonton, AB, Canada
| | - Simonetta Sipione
- Department of Pharmacology, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - John S Klassen
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Ratmir Derda
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Todd L Lowary
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
- Institute of Biological Chemistry, Academia Sinica, Nangang, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Lara K Mahal
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Meghan R Riddell
- Department of Obstetrics & Gynaecology and Physiology University of Alberta, Edmonton, AB, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada.
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
19
|
Raïch-Regué D, Resa-Infante P, Gallemí M, Laguia F, Muñiz-Trabudua X, Muñoz-Basagoiti J, Perez-Zsolt D, Chojnacki J, Benet S, Clotet B, Martinez-Picado J, Izquierdo-Useros N. Role of Siglecs in viral infections: A double-edged sword interaction. Mol Aspects Med 2023; 90:101113. [PMID: 35981912 PMCID: PMC9923124 DOI: 10.1016/j.mam.2022.101113] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/21/2023]
Abstract
Sialic-acid-binding immunoglobulin-like lectins are cell surface immune receptors known as Siglecs that play a paramount role as modulators of immunity. In recent years, research has underscored how the underlaying biology of this family of receptors influences the outcome of viral infections. While Siglecs are needed to promote effective antiviral immune responses, they can also pave the way to viral dissemination within tissues. Here, we review how recent preclinical findings focusing on the interplay between Siglecs and viruses may translate into promising broad-spectrum therapeutic interventions or key biomarkers to monitor the course of viral infections.
Collapse
Affiliation(s)
- Dàlia Raïch-Regué
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Patricia Resa-Infante
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
| | - Marçal Gallemí
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Fernando Laguia
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Xabier Muñiz-Trabudua
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | | | - Daniel Perez-Zsolt
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Jakub Chojnacki
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain
| | - Susana Benet
- Fundació lluita contra la SIDA, Infectious Diseases Department, Hospital Germans Trias i Pujol, 08916, Badalona, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Fundació lluita contra la SIDA, Infectious Diseases Department, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Javier Martinez-Picado
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain; Catalan Institution for Research and Advanced Studies (ICREA), 08010, Barcelona, Spain
| | - Nuria Izquierdo-Useros
- IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, 08916, Badalona, Spain; Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916, Badalona, Spain; Consorcio Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
20
|
Cao Y, Song W, Chen X. Multivalent sialic acid materials for biomedical applications. Biomater Sci 2023; 11:2620-2638. [PMID: 36661319 DOI: 10.1039/d2bm01595a] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Sialic acid is a kind of monosaccharide expressed on the non-reducing end of glycoproteins or glycolipids. It acts as a signal molecule combining with its natural receptors such as selectins and siglecs (sialic acid-binding immunoglobulin-like lectins) in intercellular interactions like immunological surveillance and leukocyte infiltration. The last few decades have witnessed the exploration of the roles that sialic acid plays in different physiological and pathological processes and the use of sialic acid-modified materials as therapeutics for related diseases like immune dysregulation and virus infection. In this review, we will briefly introduce the biomedical function of sialic acids in organisms and the utilization of multivalent sialic acid materials for targeted drug delivery as well as therapeutic applications including anti-inflammation and anti-virus.
Collapse
Affiliation(s)
- Yusong Cao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China. .,University of Science and Technology of China, Hefei, 230026, China.,Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, China
| |
Collapse
|
21
|
Jiang KY, Qi LL, Liu XB, Wang Y, Wang L. Prognostic value of Siglec-15 expression in patients with solid tumors: A meta-analysis. Front Oncol 2023; 12:1073932. [PMID: 36713548 PMCID: PMC9875589 DOI: 10.3389/fonc.2022.1073932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023] Open
Abstract
Background Siglec-15 is expressed in a variety of cancers. However, the role of Siglec-15 in the prognosis of cancer patients remains controversial. Therefore, we conducted a meta-analysis to clarify the potential prognostic value of Siglec-15 in solid tumors. Methods The PubMed, Web of Science, Embase and CNKI databases were comprehensively searched to identify studies assessing the effect of Siglec-15 on the survival of cancer patients. Hazard ratios (HRs) with 95% confidence intervals (CIs) for overall survival (OS), progression-free survival (PFS) and disease-specific survival (DSS) from individual studies were evaluated. Results The data from 13 observational studies consisting of 1376 patients were summarized. Elevated baseline Siglec-15 expression was significantly correlated with poor OS (pooled HR = 1.28, 95% CI: 1.05-1.56; P = 0.013). However, high Siglec-15 expression predicted a significantly better DSS (pooled HR = 0.73 (95% CI: 0.57-0.94; P = 0.015) but not PFS (pooled HR = 1.49, 95% CI: 0.46-4.87; P=0.510). In addition, high Siglec-15 expression was not associated with PD-L1 (OR=0.64, 95% CI: 0.42-0.95; P = 0.028). High Siglec-15 expression was associated with male sex (OR = 1.39, 95% CI: 1.05-1.84; P = 0.022), larger tumor size (OR = 1.896, 95% CI: 1.26-2.9; P = 0.002), and advanced tumor-node-metastasis (TNM) stage (OR = 1.84; 95% CI: 1.19-2.84; P =0.006) in solid tumors. Conclusions This updated study suggested the expression of Siglec-15 is significantly associated with poor outcomes in human solid tumors, but further studies are needed to determine the prognostic value of Siglec-15 in solid tumors.
Collapse
Affiliation(s)
- Kui-Ying Jiang
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Li-Li Qi
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China,Experimental Teaching Center, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xin-Bo Liu
- Department of thoracic surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yong Wang
- Department of Academic Research, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ling Wang
- Department of Orthopedic Oncology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China,*Correspondence: Ling Wang,
| |
Collapse
|
22
|
Huang R, Zheng J, Shao Y, Zhu L, Yang T. Siglec-15 as multifunctional molecule involved in osteoclast differentiation, cancer immunity and microbial infection. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:34-41. [PMID: 36265694 DOI: 10.1016/j.pbiomolbio.2022.10.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 09/19/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
Abstract
Siglec-15 is a highly conserved member of the Siglec family, expressed on osteoclasts, a subset of myeloid cells and some cancer cells. Except for regulating osteoclast differentiation, Siglec-15 engages in immunoregulation as an immune suppressor. Siglec-15 functions as an immunosuppressive molecule in tumor-associated macrophage-mediated T cell immunity in the tumor microenvironment (TME), which makes Siglec-15 to be an emerging and promising target for normalization cancer immunotherapy. Besides, Siglec-15 interacts with sialylated pathogens and modulates host immune response against microbial pathogens by altering cytokine production and/or phagocytosis, which further broadens the underlying pathophysiological roles of Siglec-15. The fact that N-glycosylation and sialylation of Siglec-15 play a pivotal role in Siglec-15 biological function indicates that targeting certain post-translational modification may be an effective strategy for targeting Siglec-15 therapy. In-depth exploring Siglec-15 biology function is crucial for better design of Siglec-15-based therapy according to different clinical indications.
Collapse
Affiliation(s)
- Rui Huang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Department of Clinical Laboratory, Children's Hospital and Women Health Center of Shanxi, Taiyuan, China
| | - Jinxiu Zheng
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Ying Shao
- Department of Pathophysiology, Shanxi Medical University, Taiyuan, China
| | - Lei Zhu
- Department of Clinical Laboratory, Children's Hospital and Women Health Center of Shanxi, Taiyuan, China
| | - Tao Yang
- Department of Biochemistry & Molecular Biology, Shanxi Medical University, Taiyuan, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China.
| |
Collapse
|
23
|
Läubli H, Nalle SC, Maslyar D. Targeting the Siglec-Sialic Acid Immune Axis in Cancer: Current and Future Approaches. Cancer Immunol Res 2022; 10:1423-1432. [PMID: 36264237 PMCID: PMC9716255 DOI: 10.1158/2326-6066.cir-22-0366] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/08/2022] [Accepted: 09/01/2022] [Indexed: 01/10/2023]
Abstract
The sialic acid-binding immunoglobulin-like lectin (Siglec)-sialic acid immune axis is an evolutionarily conserved immunoregulatory pathway that provides a mechanism for establishing self-recognition and combatting invasive pathogens. Perturbations in the pathway lead to many immune dysregulated diseases, including autoimmunity, neurodegeneration, allergic conditions, and cancer. The purpose of this review is to provide a brief overview of the relationship between Siglecs and sialic acid as they relate to human health and disease, to consider current Siglec-based therapeutics, and to discuss new therapeutic approaches targeting the Siglec-sialic acid immune axis, with a focus on cancer.
Collapse
Affiliation(s)
- Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University, of Basel, Basel, Switzerland.,Division of Oncology, University Hospital Basel, Basel, Switzerland.,Corresponding Author: Heinz Läubli, University Hospital Basel, Petersgraben 4, Basel 4031, Switzerland. Phone: 416-1556-5212; Fax: 416-1265-5316; E-mail:
| | | | | |
Collapse
|
24
|
Shivatare SS, Shivatare VS, Wong CH. Glycoconjugates: Synthesis, Functional Studies, and Therapeutic Developments. Chem Rev 2022; 122:15603-15671. [PMID: 36174107 PMCID: PMC9674437 DOI: 10.1021/acs.chemrev.1c01032] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glycoconjugates are major constituents of mammalian cells that are formed via covalent conjugation of carbohydrates to other biomolecules like proteins and lipids and often expressed on the cell surfaces. Among the three major classes of glycoconjugates, proteoglycans and glycoproteins contain glycans linked to the protein backbone via amino acid residues such as Asn for N-linked glycans and Ser/Thr for O-linked glycans. In glycolipids, glycans are linked to a lipid component such as glycerol, polyisoprenyl pyrophosphate, fatty acid ester, or sphingolipid. Recently, glycoconjugates have become better structurally defined and biosynthetically understood, especially those associated with human diseases, and are accessible to new drug, diagnostic, and therapeutic developments. This review describes the status and new advances in the biological study and therapeutic applications of natural and synthetic glycoconjugates, including proteoglycans, glycoproteins, and glycolipids. The scope, limitations, and novel methodologies in the synthesis and clinical development of glycoconjugates including vaccines, glyco-remodeled antibodies, glycan-based adjuvants, glycan-specific receptor-mediated drug delivery platforms, etc., and their future prospectus are discussed.
Collapse
Affiliation(s)
- Sachin S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Vidya S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
25
|
Zheng B, Song K, Sun L, Gao Y, Qu Y, Ren C, Yan P, Chen W, Guo W, Zhou C, Yue B. Siglec-15-induced autophagy promotes invasion and metastasis of human osteosarcoma cells by activating the epithelial-mesenchymal transition and Beclin-1/ATG14 pathway. Cell Biosci 2022; 12:109. [PMID: 35842729 PMCID: PMC9287887 DOI: 10.1186/s13578-022-00846-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/06/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Pulmonary metastasis is the main cause of poor prognosis in osteosarcoma. Sialic acid-bound immunoglobulin lectin 15 (Siglec-15) has been demonstrated to be obviously correlated with pulmonary metastasis in osteosarcoma patients. However, the effect of Siglec-15 on autophagy in osteosarcoma remains unclear, while the role and mechanism of Siglec-15-related autophagy in lung metastasis also remain unknown. METHODS The expression levels of Siglec-15 and Beclin-1 were detected in osteosarcoma tissues using immunohistochemistry (IHC). The effect of Siglec-15 on metastasis was investigated using Transwell, wound healing and animal experiments with osteosarcoma cells. Corresponding proteins were confirmed using Western blotting when Siglec-15 or Beclin-1 was silenced or overexpressed. Changes in autophagy and the cytoskeleton were detected using immunofluorescence and transmission electron microscopy. RESULTS Siglec-15 and Beclin-1 expression was evaluated both in lung metastases and in patients who presented with pulmonary metastasis of osteosarcoma. Immunoprecipitation experiments revealed that Siglec-15 interacts directly with Beclin-1, an important autophagic protein. Moreover, loss of Siglec-15 distinctly inhibited autophagy and reduced Beclin-1/ATG14 expression. The decreased invasion and migration caused by Siglec-15 silencing could be reversed by Beclin-1 overexpression. Additionally, autophagy can promote the epithelial-mesenchymal transition (EMT) and affect cytoskeletal rearrangement, which was confirmed by overexpression or silencing of Beclin-1. CONCLUSIONS These findings confirmed the role of Siglec-15 in the regulation of autophagy and elaborated the relationship and mechanisms between autophagy and the metastasis of osteosarcoma cells.
Collapse
Affiliation(s)
- Bingxin Zheng
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Keliang Song
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Lingling Sun
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yang Gao
- Medical Department, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yan Qu
- Industrial Investment Department, Haier, Qingdao, People's Republic of China
| | - Chongmin Ren
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Peng Yan
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China
| | - Wenfang Chen
- Department of Physiology, Medical College of Qingdao University, Qingdao, People's Republic of China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China
| | - Chuanli Zhou
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China.
| | - Bin Yue
- Department of Orthopedic Oncology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266000, People's Republic of China.
| |
Collapse
|
26
|
Xiao X, Peng Y, Wang Z, Zhang L, Yang T, Sun Y, Chen Y, Zhang W, Chang X, Huang W, Tian S, Feng Z, Xinhua N, Tang Q, Mao Y. A novel immune checkpoint siglec-15 antibody inhibits LUAD by modulating mφ polarization in TME. Pharmacol Res 2022; 181:106269. [PMID: 35605813 DOI: 10.1016/j.phrs.2022.106269] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Siglec-15 (S15) is a type-I transmembrane protein and is considered a new candidate of immune checkpoint inhibitor for cancer immunotherapy. METHODS In the present study, we first constructed and characterized a chimeric S15-specific monoclonal antibody (S15-4E6A). Then, the antitumor effectiveness and modulatory role of S15-4E6A in macrophages (mφs) were explored in vitro and in vivo. Finally, the underlying mechanism by which S15mAb inhibits LUAD was preliminarily explored. RESULTS The results demonstrated the successful construction of S15-4E6A, and S15-4E6A exerted an efficacious tumor-inhibitory effect on LUAD cells and xenografts. S15-4E6A could promote M1-mφ polarization while inhibiting M2-mφ polarization, both in vitro and in vivo. CONCLUSIONS S15-based immunotherapy that functions by modulating mφ polarization may be a promising strategy for the treatment of S15-positive LUAD.
Collapse
Affiliation(s)
- Xuejun Xiao
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China
| | - Yan Peng
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Zheyue Wang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Louqian Zhang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Tingting Yang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Yangyang Sun
- Department of Pathology, Changzhou No. 2 People's Hospital Affiliated with Nanjing Medical University, Changzhou, China
| | - Yufeng Chen
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Wenqing Zhang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Xinxia Chang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Wen Huang
- Department of Oncology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuning Tian
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China
| | - Zhenqing Feng
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Nabi Xinhua
- Department of Pharmacology, Xinjiang Medical University, Urumqi, China.
| | - Qi Tang
- NHC Key Laboratory of Antibody Technique, Nanjing Medical University, Nanjing, China; Jiangsu Province Engineering Research Center of Antibody Drug, Nanjing Medical University, Nanjing, China; Department of Pathology, Changzhou No. 2 People's Hospital Affiliated with Nanjing Medical University, Changzhou, China.
| | - Yuan Mao
- Department of Oncology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Oncology, Geriatric Hospital of Nanjing Medical University, Nanjing, China; Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China.
| |
Collapse
|
27
|
Siglec-15 Silencing Inhibits Cell Proliferation and Promotes Cell Apoptosis by Inhibiting STAT1/STAT3 Signaling in Anaplastic Thyroid Carcinoma. DISEASE MARKERS 2022; 2022:1606404. [PMID: 35769818 PMCID: PMC9236774 DOI: 10.1155/2022/1606404] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/10/2022] [Indexed: 11/17/2022]
Abstract
Thyroid cancer (THCA) represents a frequently seen endocrine cancer, which can be divided as anaplastic thyroid carcinoma (ATC), follicular thyroid carcinoma (FTC), and papillary thyroid carcinoma (PTC). A total of 362 IDEGs were obtained from TCGA-THCA and IMMPORT databases, which were found to be related to BP, CC, MF, and STAT signaling pathway upon GO functional annotation and KEGG analysis. This work identified 23 survival-related hub genes using WGCNA and uniCOX analysis. In addition, a risk prognosis model was constructed to obtain a signature involving fifteen IDEGs. According to survival and univariate along with multivariate analysis, high-risk patients had markedly dismal prognostic outcome compared with low-risk counterparts. Siglec-15 belongs to one of the fifteen IDEG signature, but the precise biological roles in diverse THCA subtypes are largely unclear. In this work, Siglec-15 expression evidently increased in ATC and FTC samples compared with matched surrounding PTC and THCA samples, which was used as a diagnostic biomarker for THCA. Siglec-15 RNAi significantly inhibited cell proliferation and promoted cell apoptosis. Meanwhile, Siglec-15 knockout suppressed the expression of STAT1, STAT3, and VEGF and promoted that of cleaved caspase-3. In vivo experiments revealed that transfection with vectors expressing STAT1 and STAT3 inhibited the Siglec-15 RNAi-induced inhibition on tumor growth and the increases in CD4+/CD8+ ratio. In conclusion, Siglec-15 expression increases in ATC and FTC, which promotes THCA occurrence via the STAT1/STAT3 signaling, in particular for FTC and ATC. Therefore, it is the possible marker that can be used to diagnose and treat THCA.
Collapse
|
28
|
Jiang KY, Qi LL, Kang FB, Wang L. The intriguing roles of Siglec family members in the tumor microenvironment. Biomark Res 2022; 10:22. [PMID: 35418152 PMCID: PMC9008986 DOI: 10.1186/s40364-022-00369-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/25/2022] [Indexed: 02/06/2023] Open
Abstract
Sialic acid-binding receptors are expressed on the surfaces of a variety of immune cells and have complex and diverse immunoregulatory functions in health and diseases. Recent studies have shown that Siglecs could play diverse immune and nonimmune regulatory roles in the tumor microenvironment (TME) and participate in tumor progression through various mechanisms, such as regulating tumor growth and metastasis, mediating the inflammatory response, and promoting tumor immune escape, thereby affecting the prognoses and outcomes of patients. However, depending on the cell type in which they are expressed, each Siglec member binds to corresponding ligands in the microenvironment milieu to drive diverse cell physiological and pathological processes in tumors. Therefore, we herein summarize the expression spectra and functions of the Siglec family in human diseases, particularly cancer, and highlight the possibility of therapeutic interventions targeting the TME in the future.
Collapse
Affiliation(s)
- Kui-Ying Jiang
- Department of Orthopedic Oncology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Li-Li Qi
- Experimental Center for Teaching of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Fu-Biao Kang
- The Liver Disease Center of PLA, the 980Th Hospital of PLA Joint Logistics Support Force, Shijiazhuang, Hebei, People's Republic of China.
| | - Ling Wang
- Department of Orthopedic Oncology, the Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
29
|
Sugar nucleotide regeneration system for the synthesis of Bi- and triantennary N-glycans and exploring their activities against siglecs. Eur J Med Chem 2022; 232:114146. [DOI: 10.1016/j.ejmech.2022.114146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/17/2022] [Accepted: 01/17/2022] [Indexed: 11/18/2022]
|
30
|
Expression of Immune Checkpoints in Malignant Tumors: Therapy Targets and Biomarkers for the Gastric Cancer Prognosis. Diagnostics (Basel) 2021; 11:diagnostics11122370. [PMID: 34943606 PMCID: PMC8700640 DOI: 10.3390/diagnostics11122370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
To increase the effectiveness of anticancer therapy based on immune checkpoint (IC) inhibition, some ICs are being investigated in addition to those used in clinic. We reviewed data on the relationship between PD-L1, B7-H3, B7-H4, IDO1, Galectin-3 and -9, CEACAM1, CD155, Siglec-15 and ADAM17 expression with cancer development in complex with the results of clinical trials on their inhibition. Increased expression of the most studied ICs—PD-L1, B7-H3, and B7-H4—is associated with poor survival; their inhibition is clinically significant. Expression of IDO1, CD155, and ADAM17 is also associated with poor survival, including gastric cancer (GC). The available data indicate that CD155 and ADAM17 are promising targets for immune therapy. However, the clinical trials of anti-IDO1 antibodies have been unsatisfactory. Expression of Galectin-3 and -9, CEACAM1 and Siglec-15 demonstrates a contradictory relationship with patient survival. The lack of satisfactory results of these IC inhibitor clinical trials additionally indicates the complex nature of their functioning. In conclusion, in many cases it is important to analyze the expression of other participants of the immune response besides target IC. The PD-L1, B7-H3, B7-H4, IDO1 and ADAM17 may be considered as candidates for prognosis markers for GC patient survival.
Collapse
|
31
|
He F, Wang N, Li J, He L, Yang Z, Lu J, Xiong G, Yu C, Wang S. High affinity monoclonal antibody targeting Siglec-15 for cancer immunotherapy. J Clin Transl Res 2021; 7:739-749. [PMID: 34988324 PMCID: PMC8710358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/11/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND AIM Recently, Siglec-15 has been proved as a novel immune suppressor and a potential target for normalization cancer immunotherapy, which is non-redundant to the well-known PD-L1/PD-1 pathway. Herein, anti-Siglec-15 mAb, a monoclonal antibody (mAb) with a high affinity against Siglec-15, was prepared. METHODS The engineered CHO-K1 Siglec-15 cell line was constructed to heterologously expressed Siglec-15 for the affinity test with the mAb. Antigens Siglec-15-mIgG and Siglec-15-his were recombinantly expressed by 293F cells and purified by high-performance liquid chromatography (HPLC). Hybridoma cell line against Siglec-15 was prepared and validated by enzyme-linked immunoabsorbant assay (ELISA) and fluorescent-activated cell sorting (FACS). Finally, the anti-Siglec-15 mAb was produced, purified, and confirmed by SDS-PAGE, ELISA, and FACS. RESULTS The EC50 of the anti-Siglec-15 mAb with Siglec-15 is 76.65 ng/mL, lower than that of the positive control 5G12 (90.7 ng/mL), indicating a high affinity of the anti-Siglec-15 mAb. In vitro and in vivo studies verified that the anti-Siglec-15 mAb blocks the Siglec-15-mediated suppression of T cell and moderately prevents the tumor growth. CONCLUSIONS The anti-Siglec-15 mAb can be considered as an effective immunotherapy for tumor suppression. RELEVANCE FOR PATIENTS The anti-Siglec-15 mAb prepared in this study is useful as an immune checkpoint inhibitor against Siglec-15 for normalization cancer immunotherapy. This immunotherapy provides an alternative treatment for cancer patients who are refractory to the well-known PD-L1/PD-1-targeting therapies.
Collapse
Affiliation(s)
- Fei He
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Na Wang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jiangwei Li
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Luanying He
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zhao Yang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,2College of Life Science, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin of Xinjiang Production and Construction Corps, Tarim University, Alar 843300, Xinjiang, China
| | - Jiandong Lu
- 3Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518033, Guangdong, China
| | - Guoliang Xiong
- 3Shenzhen Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518033, Guangdong, China
| | - Changyuan Yu
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,Corresponding authors: Changyuan Yu and Shihui Wang College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China Tel: +86-10-64421335 Fax: +86-10-64421335 E-mail: ;
| | - Shihui Wang
- 1College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China,Corresponding authors: Changyuan Yu and Shihui Wang College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China Tel: +86-10-64421335 Fax: +86-10-64421335 E-mail: ;
| |
Collapse
|
32
|
Ronit A, Jørgensen SE, Roed C, Eriksson R, Iepsen UW, Plovsing RR, Storgaard M, Gustafsson F, Hansen ABE, Mogensen TH. Host Genetics and Antiviral Immune Responses in Adult Patients With Multisystem Inflammatory Syndrome. Front Immunol 2021; 12:718744. [PMID: 34531865 PMCID: PMC8439578 DOI: 10.3389/fimmu.2021.718744] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/12/2021] [Indexed: 01/31/2023] Open
Abstract
COVID-19 associated multisystem inflammatory syndrome (MIS) is a rare condition mostly affecting children but also adults (MIS-A). Although severe systemic inflammation and multiorgan dysfunction are hallmarks of the syndrome, the underlying pathogenesis is unclear. We aimed to provide novel immunological and genetic descriptions of MIS-A patients. Cytokine responses (IL-6, IL-1β, TNFα, CXCL10, type I, II and III interferons) following SARS-CoV-2 infection of peripheral blood mononuclear cells in vitro were analyzed as well as antibodies against IFNα and IFNω (by ELISA) in patients and healthy controls. We also performed whole exome sequencing (WES) of patient DNA. A total of five patients (ages 19, 23, 33, 38, 50 years) were included. The patients shared characteristic features, although organ involvement and the time course of disease varied slightly. SARS-CoV-2 in vitro infection of patient PBMCs revealed impaired type I and III interferon responses and reduced CXCL10 expression, whereas production of proinflammatory cytokines were less affected, compared to healthy controls. Presence of interferon autoantibodies was not detected. Whole exome sequencing analysis of patient DNA revealed 12 rare potentially disease-causing variants in genes related to autophagy, classical Kawasaki disease, restriction factors and immune responses. In conclusion, we observed an impaired production of type I and III interferons in response to SARS-CoV-2 infection and detected several rare potentially disease-causing gene variants potentially contributing to MIS-A.
Collapse
Affiliation(s)
- Andreas Ronit
- Department of Infectious Diseases 144, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Sofie E Jørgensen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Casper Roed
- Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Robert Eriksson
- Department of Infectious Diseases 8632, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Department of Disease Systems Biology, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Ulrik W Iepsen
- Department of Anaesthesiology and Intensive Care, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Ronni R Plovsing
- Department of Anaesthesiology and Intensive Care, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Merete Storgaard
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Finn Gustafsson
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark.,Department of Cardiology and Clinical Medicine, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Brit E Hansen
- Department of Infectious Diseases 144, Hvidovre Hospital, University of Copenhagen, Hvidovre, Denmark
| | - Trine H Mogensen
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
33
|
Liu W, Ji Z, Wu B, Huang S, Chen Q, Chen X, Wei Y, Jiang J. Siglec-15 promotes the migration of liver cancer cells by repressing lysosomal degradation of CD44. FEBS Lett 2021; 595:2290-2302. [PMID: 34328657 DOI: 10.1002/1873-3468.14169] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 06/14/2021] [Accepted: 07/14/2021] [Indexed: 01/25/2023]
Abstract
Sialic acid-binding immunoglobulin-like lectin-15 (Siglec-15) has been identified as a novel potential target for cancer immunotherapy. Here, we explored the role of Siglec-15 in human hepatoma cells. In this study, we found that the expression of Siglec-15 is substantially upregulated in liver cancer tissues in comparison with the nontumor tissues. Functionally, in vitro experiments show that Siglec-15 promotes the migration of hepatoma cells. Furthermore, the data demonstrated an interaction between Siglec-15 and CD44, a transmembrane glycoprotein that mediates tumor progression and metastasis. In addition, we show that CD44 is modified by α2,6-linked sialic acids on N-glycans in hepatoma cells and that CD44 sialylation affects its interaction with Siglec-15. Removal of the sialic acid residues from CD44 resulted in suppressed interaction between Siglec-15 and CD44. We further demonstrate that Siglec-15 interacts and promotes the stability of CD44 by preventing its lysosomal-mediated degradation. Taken together, our findings demonstrate that Siglec-15 promotes the migration of hepatoma cells by regulating the CD44 protein stability.
Collapse
Affiliation(s)
- Weitao Liu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhi Ji
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - BingRui Wu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Sijing Huang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qihang Chen
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoning Chen
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yuanyan Wei
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianhai Jiang
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
34
|
Dufrançais O, Mascarau R, Poincloux R, Maridonneau-Parini I, Raynaud-Messina B, Vérollet C. Cellular and molecular actors of myeloid cell fusion: podosomes and tunneling nanotubes call the tune. Cell Mol Life Sci 2021; 78:6087-6104. [PMID: 34296319 PMCID: PMC8429379 DOI: 10.1007/s00018-021-03875-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/25/2021] [Accepted: 06/05/2021] [Indexed: 12/22/2022]
Abstract
Different types of multinucleated giant cells (MGCs) of myeloid origin have been described; osteoclasts are the most extensively studied because of their importance in bone homeostasis. MGCs are formed by cell-to-cell fusion, and most types have been observed in pathological conditions, especially in infectious and non-infectious chronic inflammatory contexts. The precise role of the different MGCs and the mechanisms that govern their formation remain poorly understood, likely due to their heterogeneity. First, we will introduce the main populations of MGCs derived from the monocyte/macrophage lineage. We will then discuss the known molecular actors mediating the early stages of fusion, focusing on cell-surface receptors involved in the cell-to-cell adhesion steps that ultimately lead to multinucleation. Given that cell-to-cell fusion is a complex and well-coordinated process, we will also describe what is currently known about the evolution of F-actin-based structures involved in macrophage fusion, i.e., podosomes, zipper-like structures, and tunneling nanotubes (TNT). Finally, the localization and potential role of the key fusion mediators related to the formation of these F-actin structures will be discussed. This review intends to present the current status of knowledge of the molecular and cellular mechanisms supporting multinucleation of myeloid cells, highlighting the gaps still existing, and contributing to the proposition of potential disease-specific MGC markers and/or therapeutic targets.
Collapse
Affiliation(s)
- Ophélie Dufrançais
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Rémi Mascarau
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina
| | - Renaud Poincloux
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Isabelle Maridonneau-Parini
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Toulouse, France.
- International Associated Laboratory (LIA) CNRS "IM-TB/HIV" (1167), Buenos Aires, Argentina.
| |
Collapse
|
35
|
Matsusaka K, Fujiwara Y, Pan C, Esumi S, Saito Y, Bi J, Nakamura Y, Mukunoki A, Takeo T, Nakagata N, Yoshii D, Fukuda R, Nagasaki T, Tanaka R, Komori H, Maeda H, Watanabe H, Tamada K, Komohara Y, Maruyama T. α 1-Acid Glycoprotein Enhances the Immunosuppressive and Protumor Functions of Tumor-Associated Macrophages. Cancer Res 2021; 81:4545-4559. [PMID: 34210751 DOI: 10.1158/0008-5472.can-20-3471] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 05/04/2021] [Accepted: 06/29/2021] [Indexed: 11/16/2022]
Abstract
Blood levels of acute-phase protein α1-acid glycoprotein (AGP, orosmucoid) increase in patients with cancer. Although AGP is produced from hepatocytes following stimulation by immune cell-derived cytokines under conditions of inflammation and tumorigenesis, the functions of AGP in tumorigenesis and tumor progression remain unknown. In the present study, we revealed that AGP contributes directly to tumor development by induction of programmed death ligand 1 (PD-L1) expression and IL6 production in macrophages. Stimulation of AGP induced PD-L1 expression in both human monocyte-derived macrophages through STAT1 activation, whereas AGP had no direct effect on PD-L1 expression in tumor cells. AGP also induced IL6 production from macrophages, which stimulated proliferation in tumor cells by IL6R-mediated activation of STAT3. Furthermore, administration of AGP to AGP KO mice phenocopied effects of tumor-associated macrophages (TAM) on tumor progression. AGP decreased IFNγ secretion from T cells and enhanced STAT3 activation in subcutaneous tumor tissues. In addition, AGP regulated PD-L1 expression and IL6 production in macrophages by binding with CD14, a coreceptor for Toll-like receptor 4 (TLR4), and inducing TLR4 signaling. These results provide the first evidence that AGP is directly involved in tumorigenesis by interacting with TAMs and that AGP might be a target molecule for anticancer therapy. SIGNIFICANCE: AGP-mediated suppression of antitumor immunity contributes to tumor progression by inducing PD-L1 expression and IL6 production in TAMs.
Collapse
Affiliation(s)
- Kotaro Matsusaka
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.,Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yukio Fujiwara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Cheng Pan
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Shigeyuki Esumi
- Department of Anatomy and Neurobiology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoichi Saito
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jing Bi
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuka Nakamura
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ayumi Mukunoki
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Daiki Yoshii
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ryo Fukuda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Taisei Nagasaki
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Ryusei Tanaka
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisakazu Komori
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hitoshi Maeda
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Watanabe
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Tamada
- Department of Immunology, Graduate School of Medicine, Faculty of Medicine and Health Sciences, Yamaguchi University, Yamaguchi Prefecture, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan. .,Center for Metabolic Regulation of Healthy Aging, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Maruyama
- Department of Biopharmaceutics, Graduate School of Pharmaceutical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
36
|
Yang D, Yang L, Cai J, Hu X, Li H, Zhang X, Zhang X, Chen X, Dong H, Nie H, Li Y. A sweet spot for macrophages: Focusing on polarization. Pharmacol Res 2021; 167:105576. [PMID: 33771700 DOI: 10.1016/j.phrs.2021.105576] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/21/2022]
Abstract
Macrophages are a type of functionally plastic cells that can create a pro-/anti-inflammatory microenvironment for organs by producing different kinds of cytokines, chemokines, and growth factors to regulate immunity and inflammatory responses. In addition, they can also be induced to adopt different phenotypes in response to extracellular and intracellular signals, a process defined as M1/M2 polarization. Growing evidence indicates that glycobiology is closely associated with this polarization process. In this research, we review studies of the roles of glycosylation, glucose metabolism, and key lectins in the regulation of macrophages function and polarization to provide a new perspective for immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110000, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaxin Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaohan Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xinghe Chen
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haiyang Dong
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
37
|
Cancer vaccines: An unkept promise? Drug Discov Today 2021; 26:1347-1352. [PMID: 33601016 DOI: 10.1016/j.drudis.2021.02.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/16/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Two decades ago, cancer vaccines were hailed as a prominent breakthrough for the treatment of cancer. However, the vaccines failed to show any improvement in median survival time in various clinical trials, even though they stimulated the immune response and showed exceptional safety profiles. The resistance of cancer cells to the immune response was revealed as a significant hurdle. In this review, I discuss the different types of cancer vaccines and the strategies used to design them. I also highlight how cancer cells develop resistance to the immune response, and how therapies, such as monoclonal antibodies (mAbs) and small interfering (si)RNA/short hairpin (sh)RNA could be used to address some of the shortcomings of cancer vaccine treatments.
Collapse
|
38
|
Murugesan G, Correia VG, Palma AS, Chai W, Li C, Feizi T, Martin E, Laux B, Franz A, Fuchs K, Weigle B, Crocker PR. Siglec-15 recognition of sialoglycans on tumor cell lines can occur independently of sialyl Tn antigen expression. Glycobiology 2021; 31:44-54. [PMID: 32501471 PMCID: PMC7799145 DOI: 10.1093/glycob/cwaa048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/21/2020] [Accepted: 06/01/2020] [Indexed: 12/15/2022] Open
Abstract
Siglec-15 is a conserved sialic acid-binding Ig-like lectin expressed on osteoclast progenitors, which plays an important role in osteoclast development and function. It is also expressed by tumor-associated macrophages and by some tumors, where it is thought to contribute to the immunosuppressive microenvironment. It was shown previously that engagement of macrophage-expressed Siglec-15 with tumor cells expressing its ligand, sialyl Tn (sTn), triggered production of TGF-β. In the present study, we have further investigated the interaction between Siglec-15 and sTn on tumor cells and its functional consequences. Based on binding assays with lung and breast cancer cell lines and glycan-modified cells, we failed to see evidence for recognition of sTn by Siglec-15. However, using a microarray of diverse, structurally defined glycans, we show that Siglec-15 binds with higher avidity to sialylated glycans other than sTn or related antigen sequences. In addition, we were unable to demonstrate enhanced TGF-β secretion following co-culture of Siglec-15-expressing monocytic cell lines with tumor cells expressing sTn or following Siglec-15 cross-linking with monoclonal antibodies. However, we did observe activation of the SYK/MAPK signaling pathway following antibody cross-linking of Siglec-15 that may modulate the functional activity of macrophages.
Collapse
Affiliation(s)
- Gavuthami Murugesan
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| | - Viviana G Correia
- Applied Molecular Biosciences Unit, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| | - Angelina S Palma
- Applied Molecular Biosciences Unit, Department of Chemistry, Faculty of Science and Technology, NOVA University of Lisbon, Lisbon, Portugal
| | - Wengang Chai
- Glycosciences Laboratory, Imperial College London, London, United Kingdom
| | - Chunxia Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy and Shandong Provincial Key laboratory of Glycoscience and Glycoengineering, Ocean University of China, Qingdao 266003, China
| | - Ten Feizi
- Glycosciences Laboratory, Imperial College London, London, United Kingdom
| | - Eva Martin
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397 Biberach/Riss, Germany
| | - Brigitte Laux
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397 Biberach/Riss, Germany
| | - Alexandra Franz
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397 Biberach/Riss, Germany
| | - Klaus Fuchs
- Biotherapeutics Discovery, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397 Biberach/Riss, Germany
| | - Bernd Weigle
- Cancer Immunology & Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co KG, Birkendorfer Str. 65, 88397 Biberach/Riss, Germany
| | - Paul R Crocker
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, United Kingdom
| |
Collapse
|
39
|
Current Status on Therapeutic Molecules Targeting Siglec Receptors. Cells 2020; 9:cells9122691. [PMID: 33333862 PMCID: PMC7765293 DOI: 10.3390/cells9122691] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/09/2020] [Accepted: 12/12/2020] [Indexed: 12/15/2022] Open
Abstract
The sialic acid-binding immunoglobulin-type of lectins (Siglecs) are receptors that recognize sialic acid-containing glycans. In the majority of the cases, Siglecs are expressed on immune cells and play a critical role in regulating immune cell signaling. Over the years, it has been shown that the sialic acid-Siglec axis participates in immunological homeostasis, and that any imbalance can trigger different pathologies, such as autoimmune diseases or cancer. For all this, different therapeutics have been developed that bind to Siglecs, either based on antibodies or being smaller molecules. In this review, we briefly introduce the Siglec family and we compile a description of glycan-based molecules and antibody-based therapies (including CAR-T and bispecific antibodies) that have been designed to therapeutically targeting Siglecs.
Collapse
|
40
|
Korn MA, Schmitt H, Angermüller S, Chambers D, Seeling M, Lux UT, Brey S, Royzman D, Brückner C, Popp V, Percivalle E, Bäuerle T, Zinser E, Winkler TH, Steinkasserer A, Nimmerjahn F, Nitschke L. Siglec-15 on Osteoclasts Is Crucial for Bone Erosion in Serum-Transfer Arthritis. THE JOURNAL OF IMMUNOLOGY 2020; 205:2595-2605. [PMID: 33020147 DOI: 10.4049/jimmunol.2000472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 09/07/2020] [Indexed: 11/19/2022]
Abstract
Siglec-15 is a conserved sialic acid-binding Ig-like lectin, which is expressed on osteoclasts. Deficiency of Siglec-15 leads to an impaired osteoclast development, resulting in a mild osteopetrotic phenotype. The role of Siglec-15 in arthritis is still largely unclear. To address this, we generated Siglec-15 knockout mice and analyzed them in a mouse arthritis model. We could show that Siglec-15 is directly involved in pathologic bone erosion in the K/BxN serum-transfer arthritis model. Histological analyses of joint destruction provided evidence for a significant reduction in bone erosion area and osteoclast numbers in Siglec-15-/- mice, whereas the inflammation area and cartilage destruction was comparable to wild-type mice. Thus, Siglec-15 on osteoclasts has a crucial function for bone erosion during arthritis. In addition, we generated a new monoclonal anti-Siglec-15 Ab to clarify its expression pattern on immune cells. Whereas this Ab demonstrated an almost exclusive Siglec-15 expression on murine osteoclasts and hardly any other expression on various other immune cell types, human Siglec-15 was more broadly expressed on human myeloid cells, including human osteoclasts. Taken together, our findings show a role of Siglec-15 as a regulator of pathologic bone resorption in arthritis and highlight its potential as a target for future therapies, as Siglec-15 blocking Abs are available.
Collapse
Affiliation(s)
- Marina A Korn
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Heike Schmitt
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Sieglinde Angermüller
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - David Chambers
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Michaela Seeling
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Uwe T Lux
- Division of Animal Physiology, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Stefanie Brey
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, University Hospital Erlangen, 91054 Erlangen, Germany; and
| | - Christin Brückner
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Vanessa Popp
- Preclinical Imaging Center Erlangen, Institute of Radiology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Elena Percivalle
- Preclinical Imaging Center Erlangen, Institute of Radiology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Tobias Bäuerle
- Preclinical Imaging Center Erlangen, Institute of Radiology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, University Hospital Erlangen, 91054 Erlangen, Germany; and
| | - Thomas H Winkler
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | | | - Falk Nimmerjahn
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen, 91058 Erlangen, Germany;
| |
Collapse
|
41
|
Candida albicans Virulence Factors and Pathogenicity for Endodontic Infections. Microorganisms 2020; 8:microorganisms8091300. [PMID: 32858856 PMCID: PMC7563224 DOI: 10.3390/microorganisms8091300] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/16/2020] [Accepted: 08/19/2020] [Indexed: 02/07/2023] Open
Abstract
Candida albicans (C. albicans) is the fungus most frequently isolated from endodontic root canal infections. Although recognized by dental pulp and periradicular tissue cells that elicit immune responses, it eludes host defenses and elicits cell death. Then, C. albicans binds tooth dentin, forms biofilms, and invades dentinal tubules to resist intracanal disinfectants and endodontic treatments. Insensitive to most common medicaments, it survives sequestered within biofilms and intratubular dentin. Thus, C. albicans has been associated with cases of persistent or refractory root canal infections. Its treatment strategies may require alternative intracanal irrigants, intracanal medicaments such as chlorhexidine gel or human beta defensin-3 (HBD3), Ca-Si-based obturating materials, and microsurgical procedures.
Collapse
|
42
|
Movsisyan LD, Macauley MS. Structural advances of Siglecs: insight into synthetic glycan ligands for immunomodulation. Org Biomol Chem 2020; 18:5784-5797. [PMID: 32756649 DOI: 10.1039/d0ob01116a] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are transmembrane proteins of the immunoglobulin (Ig) superfamily predominantly expressed on the cells of our immune system. Siglecs recognize sialic acid via their terminal V-set domain. In mammals, sialic acid-terminated glycolipids and glycoproteins are the ligands of Siglecs, and the monomeric affinity of Siglecs for their sialic acid-containing ligands is weak. Significant efforts have been devoted toward the development of chemically modified sialoside ligands to target Siglecs with higher affinity and selectivity. In this review we discuss natural and synthetic sialoside ligands for each human Siglec, emphasizing the ligand binding determinants uncovered from recent advances in protein structural information. Potential therapeutic applications of these ligands are also discussed.
Collapse
Affiliation(s)
- Levon D Movsisyan
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Matthew S Macauley
- Department of Chemistry, University of Alberta, Edmonton, Alberta, Canada and Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
43
|
Sirufo MM, De Pietro F, Bassino EM, Ginaldi L, De Martinis M. Osteoporosis in Skin Diseases. Int J Mol Sci 2020; 21:E4749. [PMID: 32635380 PMCID: PMC7370296 DOI: 10.3390/ijms21134749] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022] Open
Abstract
Osteoporosis (OP) is defined as a generalized skeletal disease characterized by low bone mass and an alteration of the microarchitecture that lead to an increase in bone fragility and, therefore, an increased risk of fractures. It must be considered today as a true public health problem and the most widespread metabolic bone disease that affects more than 200 million people worldwide. Under physiological conditions, there is a balance between bone formation and bone resorption necessary for skeletal homeostasis. In pathological situations, this balance is altered in favor of osteoclast (OC)-mediated bone resorption. During chronic inflammation, the balance between bone formation and bone resorption may be considerably affected, contributing to a net prevalence of osteoclastogenesis. Skin diseases are the fourth cause of human disease in the world, affecting approximately one third of the world's population with a prevalence in elderly men. Inflammation and the various associated cytokine patterns are the basis of both osteoporosis and most skin pathologies. Moreover, dermatological patients also undergo local or systemic treatments with glucocorticoids and immunosuppressants that could increase the risk of osteoporosis. Therefore, particular attention should be paid to bone health in these patients. The purpose of the present review is to take stock of the knowledge in this still quite unexplored field, despite the frequency of such conditions in clinical practice.
Collapse
Affiliation(s)
- Maria Maddalena Sirufo
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (E.M.B.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 64100 Teramo, Italy
| | - Francesca De Pietro
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (E.M.B.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 64100 Teramo, Italy
| | - Enrica Maria Bassino
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (E.M.B.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 64100 Teramo, Italy
| | - Lia Ginaldi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (E.M.B.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 64100 Teramo, Italy
| | - Massimo De Martinis
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (M.M.S.); (F.D.P.); (E.M.B.); (L.G.)
- Allergy and Clinical Immunology Unit, Center for the Diagnosis and Treatment of Osteoporosis, AUSL 04 64100 Teramo, Italy
| |
Collapse
|