1
|
Qiao Y, Tian X, Li S, Niu H. Identification and experimental validation of a sialylation-related long noncoding RNA signature for prognosis of bladder cancer. BMC Urol 2024; 24:222. [PMID: 39390546 PMCID: PMC11465731 DOI: 10.1186/s12894-024-01613-6] [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: 02/02/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND The dysregulation of sialylation plays a pivotal role in cancer progression and metastasis, impacting various aspects of tumor behavior. This study aimed to investigate the prognostic significance of long non-coding RNAs (lncRNAs) in relation to sialylation. Additionally, we aimed to develop a signature of sialylation-related lncRNAs in the context of bladder cancer. METHODS This study used transcriptomic data and clinical information from the TCGA (the Cancer Genome Atlas) database to screen for sialylation-related lncRNAs and constructed a prognostic model. The relationships between these lncRNAs and biological pathways, immune cell infiltration, drug sensitivity, etc., were analyzed, and the expression of some lncRNAs was validated at the cellular level. RESULTS This study identified 6 prognostic lncRNAs related to sialylation and constructed a risk score model with high predictive accuracy and reliability. The survival period of patients in the high-risk group was significantly lower than that of the low-risk group, and it was related to various biological pathways and immune functions. In addition, this study found differences in the sensitivity of patients in different risk groups to chemotherapy drugs, providing a reference for personalized treatment. CONCLUSION In this study, we examined the relationship between sialylation-related lncRNA and the prognosis of bladder cancer, providing new molecular markers and potential targets for diagnosis and treatment. Our research revealed correlations between sialylation-related lncRNA characteristics and clinicopathological features, potential mechanisms, somatic mutations, immune microenvironment, chemotherapy response, and predicted drug sensitivity in bladder cancer. Additionally, in vitro cellular studies were conducted to validate these findings and lay the groundwork for future clinical applications.
Collapse
Affiliation(s)
- Yi Qiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xintao Tian
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shengxian Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Jia Y, Lu W, Xie H, Sheng Y, Wang L, Lv W, Ling L, Dong J, Jia X, Wu S, Liu W, Ying H. Upregulation of Siglec-6 induces mitochondrial dysfunction by promoting GPR20 expression in early-onset preeclampsia. J Transl Med 2024; 22:674. [PMID: 39039496 PMCID: PMC11265165 DOI: 10.1186/s12967-024-05505-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/12/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Preeclampsia, especially early-onset preeclampsia (EO-PE), is a pregnancy complication that has serious consequences for the health of both the mother and the fetus. Although abnormal placentation due to mitochondrial dysfunction is speculated to contribute to the development of EO-PE, the underlying mechanisms have yet to be fully elucidated. METHODS The expression and localization of Siglec-6 in the placenta from normal pregnancies, preterm birth and EO-PE patients were examined by RT-qPCR, Western blot and IHC. Transwell assays were performed to evaluate the effect of Siglec-6 on trophoblast cell migration and invasion. Seahorse experiments were conducted to assess the impact of disrupting Siglec-6 expression on mitochondrial function. Co-IP assay was used to examine the interaction of Siglec-6 with SHP1/SHP2. RNA-seq was employed to investigate the mechanism by which Siglec-6 inhibits mitochondrial function in trophoblast cells. RESULTS The expression of Siglec-6 in extravillous trophoblasts is increased in placental tissues from EO-PE patients. Siglec-6 inhibits trophoblast cell migration and invasion and impairs mitochondrial function. Mechanismly, Siglec-6 inhibits the activation of NF-κB by recruiting SHP1/SHP2, leading to increased expression of GPR20. Notably, the importance of GPR20 function downstream of Siglec-6 in trophoblasts is supported by the observation that GPR20 downregulation rescues defects caused by Siglec-6 overexpression. Finally, overexpression of Siglec-6 in the placenta induces a preeclampsia-like phenotype in a pregnant mouse model. CONCLUSIONS This study indicates that the regulatory pathway Siglec-6/GPR20 has a crucial role in regulating trophoblast mitochondrial function, and we suggest that Siglec-6 and GPR20 could serve as potential markers and targets for the clinical diagnosis and therapy of EO-PE.
Collapse
Affiliation(s)
- Yuanhui Jia
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenjing Lu
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Han Xie
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yifan Sheng
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Luyao Wang
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenqi Lv
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lijun Ling
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiaqi Dong
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinrui Jia
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengyu Wu
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenqiang Liu
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Ying
- Clinical and Translational Research Center, Department of Obstetrics, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
3
|
Nunes J, Tafesse R, Mao C, Purcell M, Mo X, Zhang L, Long M, Cyr MG, Rader C, Muthusamy N. Siglec-6 as a therapeutic target for cell migration and adhesion in chronic lymphocytic leukemia. Nat Commun 2024; 15:5180. [PMID: 38890323 PMCID: PMC11189495 DOI: 10.1038/s41467-024-48678-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/08/2024] [Indexed: 06/20/2024] Open
Abstract
Siglec-6 is a lectin receptor with restricted expression in the placenta, mast cells and memory B-cells. Although Siglec-6 is expressed in patients with chronic lymphocytic leukemia (CLL), its pathophysiological role has not been elucidated. We describe here a role for Siglec-6 in migration and adhesion of CLL B cells to CLL- bone marrow stromal cells (BMSCs) in vitro and compromised migration to bone marrow and spleen in vivo. Mass spectrometry analysis revealed interaction of Siglec-6 with DOCK8, a guanine nucleotide exchange factor. Stimulation of MEC1-002 CLL cells with a Siglec-6 ligand, sTn, results in Cdc42 activation, WASP protein recruitment and F-actin polymerization, which are all associated with cell migration. Therapeutically, a Siglec-6/CD3-bispecific T-cell-recruiting antibody (T-biAb) improves overall survival in an immunocompetent mouse model and eliminates CLL cells in a patient derived xenograft model. Our findings thus reveal a migratory role for Siglec-6 in CLL, which can be therapeutically targeted using a Siglec-6 specific T-biAb.
Collapse
MESH Headings
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Humans
- Animals
- Cell Movement
- Cell Adhesion
- Lectins/metabolism
- Mice
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Female
- B-Lymphocytes/metabolism
- B-Lymphocytes/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Cell Line, Tumor
- Mesenchymal Stem Cells/metabolism
- Male
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jessica Nunes
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Rakeb Tafesse
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Molecular, Cellular and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, USA
| | - Charlene Mao
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew Purcell
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, OH, USA
| | - Liwen Zhang
- Campus Chemical Instrument Center, The Ohio State University, Columbus, OH, USA
| | - Meixiao Long
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Matthew G Cyr
- UF Scripps Biomedical Research, University of Florida, Jupiter, FL, USA
| | - Christoph Rader
- UF Scripps Biomedical Research, University of Florida, Jupiter, FL, USA
| | - Natarajan Muthusamy
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Stanczak MA, Läubli H. Siglec receptors as new immune checkpoints in cancer. Mol Aspects Med 2023; 90:101112. [PMID: 35948467 DOI: 10.1016/j.mam.2022.101112] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy in the form of immune checkpoint inhibitors and cellular therapies has improved the treatment and prognosis of many patients. Nevertheless, most cancers are still resistant to currently approved cancer immunotherapies. New approaches and rational combinations are needed to overcome these resistances. There is emerging evidence that Siglec receptors could be regarded as new immune checkpoints and targets for cancer immunotherapy. In this review, we summarize the experimental evidence supporting Siglec receptors as new immune checkpoints in cancer and discuss their mechanisms of action, as well as current efforts to target Siglec receptors and their interactions with sialoglycan Siglec-ligands.
Collapse
Affiliation(s)
- Michal A Stanczak
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, MD, 21287, USA
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy, Department of Biomedicine, University of Basel, Division of Oncology, University Hospital Basel, Switzerland.
| |
Collapse
|
5
|
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: 9.5] [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
|
6
|
Benmerzoug S, Chevalier MF, Villier L, Nguyen S, Cesson V, Schneider AK, Dartiguenave F, Rodrigues-Dias SC, Lucca I, Jichlinski P, Roth B, Nardelli-Haefliger D, Derré L. Siglec-7 May Limit Natural Killer Cell-mediated Antitumor responses in Bladder Cancer Patients. EUR UROL SUPPL 2021; 34:79-82. [PMID: 34825225 PMCID: PMC8605422 DOI: 10.1016/j.euros.2021.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2021] [Indexed: 01/03/2023] Open
Abstract
Aberrant glycosylation actively contributes to tumor progression and is a key hallmark of cancer. Most of the glycan moieties expressed on the surface of cancer cells are sialic acids that may modulate antitumor immune responses via binding to sialic acid–binding immunoglobulin-like lectins (Siglecs) expressed by immune cells. Here we show that Siglecs may decrease the bladder tumor immune response mediated by natural killer (NK) cells. We observed higher NK cell activity against desialylated bladder tumor cell lines. We therefore determined the expression of nine Siglecs on circulatory NK cells from healthy donors and patients with bladder cancer (BCa). NK cells from blood mainly express Siglec-7, which is highly upregulated in non–muscle-invasive BCa (NMIBC), as well as Siglec-6, albeit at a much lower level. However, both Siglecs are expressed by urinary NK cells from NMIBC patients undergoing bacillus Calmette-Guérin therapy. Ex vivo analysis of Siglec-6 and Siglec-7 expression levels on tumor-infiltrating NK cells (TINKs) from BCa patients showed that only Siglec-7 is expressed by TINKs. Finally, analyses for The Cancer Genome Atlas data set revealed that BCa patients with high expression levels of Siglec-7 have a poor survival rate. This work indicates that Siglec-7 may restrain NK-mediated antitumor immunity in BCa. Patient summary We investigated the expression of proteins called Siglecs in natural killer (NK) cells from patients with bladder cancer. We showed that levels of the protein Siglec-7 in blood, urine, and tumors from patients with bladder cancer are associated with poor clinical outcomes. Thus, Siglec-7 may be involved in the regulation of antitumor immunity mediated by NK cells in bladder cancer.
Collapse
Affiliation(s)
- Sulayman Benmerzoug
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Mathieu F Chevalier
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.,INSERM U976, Human Immunology, Pathophysiology and Immunotherapy Unit, Hôpital Saint-Louis, Paris, France.,Institut de Recherche Saint Louis, Université de Paris, Hôpital Saint-Louis, Paris, France
| | - Laura Villier
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sylvain Nguyen
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Valérie Cesson
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anna K Schneider
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Florence Dartiguenave
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Sonia-Christina Rodrigues-Dias
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Ilaria Lucca
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Patrice Jichlinski
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Beat Roth
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Denise Nardelli-Haefliger
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Laurent Derré
- Department of Urology, Urology Research Unit, Centre Hospitalier Universitaire Vaudois, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
7
|
Lim J, Sari-Ak D, Bagga T. Siglecs as Therapeutic Targets in Cancer. BIOLOGY 2021; 10:1178. [PMID: 34827170 PMCID: PMC8615218 DOI: 10.3390/biology10111178] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Hypersialylation is a common post-translational modification of protein and lipids found on cancer cell surfaces, which participate in cell-cell interactions and in the regulation of immune responses. Sialic acids are a family of nine-carbon α-keto acids found at the outermost ends of glycans attached to cell surfaces. Given their locations on cell surfaces, tumor cells aberrantly overexpress sialic acids, which are recognized by Siglec receptors found on immune cells to mediate broad immunomodulatory signaling. Enhanced sialylation exposed on cancer cell surfaces is exemplified as "self-associated molecular pattern" (SAMP), which tricks Siglec receptors found on leukocytes to greatly down-regulate immune responsiveness, leading to tumor growth. In this review, we focused on all 15 human Siglecs (including Siglec XII), many of which still remain understudied. We also highlighted strategies that disrupt the course of Siglec-sialic acid interactions, such as antibody-based therapies and sialic acid mimetics leading to tumor cell depletion. Herein, we introduced the central roles of Siglecs in mediating pro-tumor immunity and discussed strategies that target these receptors, which could benefit improved cancer immunotherapy.
Collapse
Affiliation(s)
- Jackwee Lim
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| | - Duygu Sari-Ak
- Department of Medical Biology, School of Medicine, University of Health Sciences, Istanbul 34668, Turkey;
| | - Tanaya Bagga
- Singapore Immunology Network, A*STAR, 8a Biomedical Grove, Singapore 138648, Singapore;
| |
Collapse
|