1
|
Aval OS, Ahmadi A, Hemid Al-Athari AJ, Soleimani Samarkhazan H, Sotudeh Chafi F, Asadi M, Mohammadi MH, Aghaei M. "Galectin-9: A double-edged sword in Acute Myeloid Leukemia". Ann Hematol 2025:10.1007/s00277-025-06387-x. [PMID: 40341460 DOI: 10.1007/s00277-025-06387-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Acute Myeloid Leukemia (AML) presents a formidable challenge in the realm of hematologic malignancies, characterized by the unregulated proliferation of myeloid progenitor cells, leading to severe disruptions in normal hematopoiesis. This review examines the multifaceted role of Galectin-9, a crucial glycan-binding protein in the pathophysiology of AML, emphasizing its potential as both a prognostic biomarker and a therapeutic target. Recent insights into the molecular underpinnings of AML, particularly those involving genetic mutations and cytogenetic abnormalities, illuminate the complex landscape of this disease, where patient outcomes are significantly influenced by individual biological markers. Galectin-9, initially recognized for its involvement in fundamental biological processes such as cell proliferation and immune modulation, has emerged as a pivotal molecule in AML, with expression levels correlating with leukemic cell behavior and clinical prognosis. This review consolidates the extensive literature on Galectin-9, elucidating its role in leukemic transformation and the therapeutic implications of manipulating this pathway. By investigating the intricate relationship between Galectin-9 and AML, we aim to provide a comprehensive understanding that could lead to innovative strategies for managing this aggressive malignancy, offering hope for improved survival outcomes through targeted therapeutic interventions.
Collapse
Affiliation(s)
- Omolbanin Sargazi Aval
- Department of Hematology, Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Ali Ahmadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sotudeh Chafi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Asadi
- HSCT research center, Bone Marrow Transplantation and Cell Therapy Lab, Taleghani hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Mohammadi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
2
|
Shil RK, Mohammed NBB, Dimitroff CJ. Galectin-9 - ligand axis: an emerging therapeutic target for multiple myeloma. Front Immunol 2024; 15:1469794. [PMID: 39386209 PMCID: PMC11461229 DOI: 10.3389/fimmu.2024.1469794] [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: 07/24/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
Galectin-9 (Gal-9) is a tandem-repeat galectin with diverse roles in immune homeostasis, inflammation, malignancy, and autoimmune diseases. In cancer, Gal-9 displays variable expression patterns across different tumor types. Its interactions with multiple binding partners, both intracellularly and extracellularly, influence key cellular processes, including immune cell modulation and tumor microenvironment dynamics. Notably, Gal-9 binding to cell-specific glycoconjugate ligands has been implicated in both promoting and suppressing tumor progression. Here, we provide insights into Gal-9 and its involvement in immune homeostasis and cancer biology with an emphasis on multiple myeloma (MM) pathophysiology, highlighting its complex and context-dependent dual functions as a pro- and anti-tumorigenic molecule and its potential implications for therapy in MM patients.
Collapse
Affiliation(s)
- Rajib K. Shil
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Norhan B. B. Mohammed
- The Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Charles J. Dimitroff
- Department of Cellular and Molecular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| |
Collapse
|
3
|
Zhang M, Liu C, Li Y, Li H, Zhang W, Liu J, Wang L, Sun C. Galectin-9 in cancer therapy: from immune checkpoint ligand to promising therapeutic target. Front Cell Dev Biol 2024; 11:1332205. [PMID: 38264357 PMCID: PMC10803597 DOI: 10.3389/fcell.2023.1332205] [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: 11/02/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Galectin-9 (Gal-9) is a vital member of the galectin family, functioning as a multi-subtype galactose lectin with diverse biological roles. Recent research has revealed that Gal-9's interaction with tumors is an independent factor that influences tumor progression. Furthermore, Gal-9 in the immune microenvironment cross-talks with tumor-associated immune cells, informing the clarification of Gal-9's identity as an immune checkpoint. A thorough investigation into Gal-9's role in various cancer types and its interaction with the immune microenvironment could yield novel strategies for subsequent targeted immunotherapy. This review focuses on the latest advances in understanding the direct and indirect cross-talk between Gal-9 and hematologic malignancies, in addition to solid tumors. In addition, we discuss the prospects of Gal-9 in tumor immunotherapy, including its cross-talk with the ligand TIM-3 and its potential in immune-combination therapy.
Collapse
Affiliation(s)
- Minpu Zhang
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cun Liu
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Ye Li
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Huayao Li
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
| | - Wenfeng Zhang
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jingyang Liu
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Liquan Wang
- Department of Thyroid and Breast Surgery, Weifang People’s Hospital, Weifang, China
| | - Changgang Sun
- College of Traditional Chinese Medicine, Weifang Medical University, Weifang, China
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang, China
| |
Collapse
|
4
|
Yang Q, Sun J, Wu W, Xing Z, Yan X, Lv X, Wang L, Song L. A galectin-9 involved in the microbial recognition and haemocyte autophagy in the Pacific oyster Crassostrea gigas. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 149:105063. [PMID: 37730190 DOI: 10.1016/j.dci.2023.105063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 09/16/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023]
Abstract
Galectin-9 is a tandem-repeat type member of galectin family participating in various immune responses, such as cell agglutination, phagocytosis, and autophagy. In the present study, a tandem repeat galectin-9 (defined as CgGal-9) was identified from Pacific oyster Crassostrea gigas, which consisted of two conserved carbohydrate recognition domains (CRDs) joined by a linker peptide. CgGal-9 was closely clustered with CaGal-9 from C. angulata, and they were assigned into the branch of invertebrate galectin-9s in the phylogenetic tree. The mRNA transcripts of CgGal-9 were detected in all the tested tissues, with the highest expression level in haemocytes. The mRNA expressions of CgGal-9 in haemocytes increased significantly after lipopolysaccharide (LPS) and Vibrio splendidus stimulation. The recombinant CgGal-9 was able to bind all the examined pathogen-associated molecular patterns (LPS, peptidoglycan, and mannose) and microbes (V. splendidus, Escherichia coli, Micrococcus luteus, Staphylococcus aureus, Bacillus subtilis, and Pichia pastoris), and agglutinated most of them in the presence of Ca2+. In CgGal-9-RNAi oysters, the mRNA expressions of autophagy related genes (CgBeclin1, CgATG5, CgP62 and CgLC3) in haemocytes decreased significantly while that of CgmTOR increased significantly at 3 h after V. splendidus stimulation. The autophagy level and mRNA expressions of autophagy related genes decreased in haemocytes after CgGal-9 was blocked by the corresponding antibody. These results revealed that CgGal-9 was able to bind different microbes and might be involved in haemocyte autophagy in the immune response of oyster.
Collapse
Affiliation(s)
- Qian Yang
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Wei Wu
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Zhen Xing
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoxue Yan
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Xiaoqian Lv
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China
| | - Lingling Wang
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Prevention and Control of Aquatic Animal Diseases, Dalian Ocean University, Dalian, 116023, China.
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, 116023, China; Southern Laboratory of Ocean Science and Engineering (Guangdong, Zhuhai), Zhuhai, 519000, China; Laboratory of Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266235, China; Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, 116023, China; Dalian Key Laboratory of Prevention and Control of Aquatic Animal Diseases, Dalian Ocean University, Dalian, 116023, China
| |
Collapse
|
5
|
Lv Y, Ma X, Ma Y, Du Y, Feng J. A new emerging target in cancer immunotherapy: Galectin-9 (LGALS9). Genes Dis 2023; 10:2366-2382. [PMID: 37554219 PMCID: PMC10404877 DOI: 10.1016/j.gendis.2022.05.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/09/2022] [Accepted: 05/14/2022] [Indexed: 11/20/2022] Open
Abstract
Over the past few decades, advances in immunological knowledge have led to the identification of novel immune checkpoints, reinvigorating cancer immunotherapy. Immunotherapy, represented by immune checkpoint inhibitors, has become the leader in the precision treatment of cancer, bringing a new dawn to the treatment of most cancer patients. Galectin-9 (LGALS9), a member of the galectin family, is a widely expressed protein involved in immune regulation and tumor pathogenesis, and affects the prognosis of various types of cancer. Galectin-9 regulates immune homeostasis and tumor cell survival through its interaction with its receptor Tim-3. In the review, based on a brief description of the signaling mechanisms and immunomodulatory activities of galectin-9 and Tim-3, we summarize the targeted expression patterns of galectin-9 in a variety of malignancies and the promising mechanisms of anti-galectin-9 therapy in stimulating anti-tumor immune responses.
Collapse
Affiliation(s)
- Yan Lv
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xiao Ma
- Department of General Surgery, The Affiliated Zhongda Hospital of Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuxin Ma
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Yuxin Du
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Jifeng Feng
- The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| |
Collapse
|
6
|
Zheng L, Xia J, Ge P, Meng Y, Li W, Li M, Wang M, Song C, Fan Y, Zhou Y. The interrelation of galectins and autophagy. Int Immunopharmacol 2023; 120:110336. [PMID: 37262957 DOI: 10.1016/j.intimp.2023.110336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 06/03/2023]
Abstract
Autophagy is a vital physiological process that maintains intracellular homeostasis by removing damaged organelles and senescent or misfolded molecules. However, excessive autophagy results in cell death and apoptosis, which will lead to a variety of diseases. Galectins are a type of animal lectin that binds to β-galactosides and can bind to the cell surface or extracellular matrix glycans, affecting a variety of immune processes in vivo and being linked to the development of many diseases. In many cases, galectins and autophagy both play important regulatory roles in the cellular life course, yet our understanding of the relationship between them is still incomplete. Galectins and autophagy may share common etiological cofactors for some diseases. Hence, we summarize the relationship between galectins and autophagy, aiming to draw attention to the existence of multiple associations between galectins and autophagy in a variety of physiological and pathological processes, which provide new ideas for etiological diagnosis, drug development, and therapeutic targets for related diseases.
Collapse
Affiliation(s)
- Lujuan Zheng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Jing Xia
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Pengyu Ge
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Yuhan Meng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Weili Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Mingming Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Min Wang
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Chengcheng Song
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Yuying Fan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| | - Yifa Zhou
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun 130024, China.
| |
Collapse
|
7
|
Morishita A, Oura K, Tadokoro T, Shi T, Fujita K, Tani J, Atsukawa M, Masaki T. Galectin-9 in Gastroenterological Cancer. Int J Mol Sci 2023; 24:ijms24076174. [PMID: 37047155 PMCID: PMC10094448 DOI: 10.3390/ijms24076174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Immunochemotherapy has become popular in recent years. The detailed mechanisms of cancer immunity are being elucidated, and new developments are expected in the future. Apoptosis allows tissues to maintain their form, quantity, and function by eliminating excess or abnormal cells. When apoptosis is inhibited, the balance between cell division and death is disrupted and tissue homeostasis is impaired. This leads to dysfunction and the accumulation of genetically abnormal cells, which can contribute to carcinogenesis. Lectins are neither enzymes nor antibodies but proteins that bind sugar chains. Among soluble endogenous lectins, galectins interact with cell surface sugar chains outside the cell to regulate signal transduction and cell growth. On the other hand, intracellular lectins are present at the plasma membrane and regulate signal transduction by regulating receptor–ligand interactions. Galectin-9 expressed on the surface of thymocytes induces apoptosis of T lymphocytes and plays an essential role in immune self-tolerance by negative selection in the thymus. Furthermore, the administration of extracellular galectin-9 induces apoptosis of human cancer and immunodeficient cells. However, the detailed pharmacokinetics of galectin-9 in vivo have not been elucidated. In addition, the cell surface receptors involved in galectin-9-induced apoptosis of cancer cells have not been identified, and the intracellular pathways involved in apoptosis have not been fully investigated. We have previously reported that galectin-9 induces apoptosis in various gastrointestinal cancers and suppresses tumor growth. However, the mechanism of galectin-9 and apoptosis induction in gastrointestinal cancers and the detailed mechanisms involved in tumor growth inhibition remain unknown. In this article, we review the effects of galectin-9 on gastrointestinal cancers and its mechanisms.
Collapse
|
8
|
Liu D, Zhu H, Li C. Galectins and galectin-mediated autophagy regulation: new insights into targeted cancer therapy. Biomark Res 2023; 11:22. [PMID: 36814341 PMCID: PMC9945697 DOI: 10.1186/s40364-023-00466-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/17/2023] [Indexed: 02/24/2023] Open
Abstract
Galectins are animal lectins with specific affinity for galactosides via the conserved carbohydrate recognition domains. Increasing studies recently have identified critical roles of galectin family members in tumor progression. Abnormal expression of galectins contributes to the proliferation, metastasis, epithelial-mesenchymal transformation (EMT), immunosuppression, radio-resistance and chemoresistance in various cancers, which has attracted cumulative clinical interest in galectin-based cancer treatment. Galectin family members have been reported to participate in autophagy regulation under physiological conditions and in non-tumoral diseases, and implication of galectins in multiple processes of carcinogenesis also involves regulation of autophagy, however, the relationship between galectins, autophagy and cancer remains largely unclear. In this review, we introduce the structure and function of galectins at the molecular level, summarize their engagements in autophagy and cancer progression, and also highlight the regulation of autophagy by galectins in cancer as well as the therapeutic potentials of galectin and autophagy-based strategies. Elaborating on the mechanism of galectin-regulated autophagy in cancers will accelerate the exploitation of galectins-autophagy targeted therapies in treatment for cancer.
Collapse
Affiliation(s)
- Dan Liu
- grid.33199.310000 0004 0368 7223Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongtao Zhu
- grid.412793.a0000 0004 1799 5032Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chuanzhou Li
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Andrzejczak A, Tupikowski K, Tomkiewicz A, Małkiewicz B, Ptaszkowski K, Domin A, Szydełko T, Karabon L. The Variations' in Genes Encoding TIM-3 and Its Ligand, Galectin-9, Influence on ccRCC Risk and Prognosis. Int J Mol Sci 2023; 24:ijms24032042. [PMID: 36768365 PMCID: PMC9917084 DOI: 10.3390/ijms24032042] [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: 11/29/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023] Open
Abstract
Renal cell cancer is the most common type of kidney cancer in adults, and clear cell renal cell carcinoma (ccRCC) is the most diagnosed type. T cell immunoglobulin and mucin-domain-containing-3 (TIM-3) belongs to immunological checkpoints that are key regulators of the immune response. One of the known TIM-3 ligands is galectin-9 (LGALS9). A limited number of studies have shown an association between TIM-3 polymorphisms and cancer risk in the Asian population; however, there is no study on the role of LGALS9 polymorphisms in cancer. The present study aimed to analyze the influence of TIM-3 and LGALS9 polymorphisms on susceptibility to ccRCC and patient overall survival (OS), with over ten years of observations. Using TaqMan probes, ARMS-PCR, and RFPL-PCR, we genotyped two TIM-3 single-nucleotide polymorphisms (SNPs): rs1036199 and rs10057302, and four LGALS9 SNPs: rs361497, rs3751093, rs4239242, and rs4794976. We found that the presence of the rs10057302 A allele (AC + AA genotypes) as well as the rs4794976 T allele (GT + TT genotypes) decreased susceptibility to ccRCC by two-fold compared to corresponding homozygotes. A subgroup analysis showed the association of some SNPs with clinical features. Moreover, TIM-3 rs1036199 significantly influenced OS. Our results indicate that variations within TIM-3 and LGALS9 genes are associated with ccRCC risk and OS.
Collapse
Affiliation(s)
- Anna Andrzejczak
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Correspondence: (A.A.); (L.K.)
| | - Krzysztof Tupikowski
- Subdivision of Urology, Lower Silesian Center for Oncology, Pulmonology and Hematology, 53-413 Wrocław, Poland
| | - Anna Tomkiewicz
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
| | - Bartosz Małkiewicz
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Kuba Ptaszkowski
- Department of Clinical Biomechanics and Physiotherapy in Motor System Disorders, Wrocław Medical University, 50-556 Wrocław, Poland
| | - Aleksandra Domin
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
| | - Tomasz Szydełko
- University Center of Excellence in Urology, Department of Minimally Invasive and Robotic Urology, Wroclaw Medical University, 50-556 Wrocław, Poland
| | - Lidia Karabon
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland
- Correspondence: (A.A.); (L.K.)
| |
Collapse
|
10
|
Godefa TM, Derks S, Thijssen VLJL. Galectins in Esophageal Cancer: Current Knowledge and Future Perspectives. Cancers (Basel) 2022; 14:5790. [PMID: 36497271 PMCID: PMC9736038 DOI: 10.3390/cancers14235790] [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: 10/12/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022] Open
Abstract
Esophageal cancer is a disease with poor overall survival. Despite advancements in therapeutic options, the treatment outcome of esophageal cancer patients remains dismal with an overall 5-year survival rate of approximately 20 percent. To improve treatment efficacy and patient survival, efforts are being made to identify the factors that underlie disease progression and that contribute to poor therapeutic responses. It has become clear that some of these factors reside in the tumor micro-environment. In particular, the tumor vasculature and the tumor immune micro-environment have been implicated in esophageal cancer progression and treatment response. Interestingly, galectins represent a family of glycan-binding proteins that has been linked to both tumor angiogenesis and tumor immunosuppression. Indeed, in several cancer types, galectins have been identified as diagnostic and/or prognostic markers. However, the role of galectins in esophageal cancer is still poorly understood. Here, we summarize the current literature with regard to the expression and potential functions of galectins in esophageal cancer. In addition, we highlight the gaps in the current knowledge and we propose directions for future research in order to reveal whether galectins contribute to esophageal cancer progression and provide opportunities to improve the treatment and survival of esophageal cancer patients.
Collapse
Affiliation(s)
- Tesfay M. Godefa
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Sarah Derks
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521 AL Utrecht, The Netherlands
| | - Victor L. J. L. Thijssen
- Cancer Center Amsterdam, Cancer Biology & Immunology, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Radiation Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
11
|
Yeung ST, Premeaux TA, Du L, Niki T, Pillai SK, Khanna KM, Ndhlovu LC. Galectin-9 protects humanized-ACE2 immunocompetent mice from SARS-CoV-2 infection. Front Immunol 2022; 13:1011185. [PMID: 36325323 PMCID: PMC9621319 DOI: 10.3389/fimmu.2022.1011185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/30/2022] [Indexed: 11/17/2022] Open
Abstract
SARS-CoV-2 remains a global health crisis even with effective vaccines and the availability of FDA approved therapies. Efforts to understand the complex disease pathology and develop effective strategies to limit mortality and morbidity are needed. Recent studies reveal circulating Galectin-9 (gal-9), a soluble beta-galactoside binding lectin with immunoregulatory properties, are elevated in SARS-CoV-2 infected individuals with moderate to severe disease. Moreover, in silico studies demonstrate gal-9 can potentially competitively bind the ACE2 receptor on susceptible host cells. Here, we determined whether early introduction of exogenous gal-9 following SARS-CoV-2 infection in humanized ACE2 transgenic mice (K18-hACE2) may reduce disease severity. Mice were infected and treated with a single dose of a human recombinant form of gal-9 (rh-gal-9) and monitored for morbidity. Subgroups of mice were humanely euthanized at 2- and 5- days post infection (dpi) for viral levels by plaque assay, immune changes measures by flow cytometry, and soluble mediators by protein analysis from lung tissue and bronchoalveolar Lavage fluid (BALF). Mice treated with rh-gal-9 during acute infection had improved survival compared to PBS treated controls. At 5 dpi, rh-gal-9 treated mice had enhanced viral clearance in the BALF, but not in the lung parenchyma. Increased T and dendritic cells and decreased neutrophil frequencies in the lung at 5 dpi were observed, whereas BALF had elevated levels of type-I interferons and proinflammatory cytokines. These results suggest a role for rh-gal-9 in limiting acute COVID-19. Further studies are required to determine the optimal design of gal-9 treatment to effectively ameliorate COVID-19 disease.
Collapse
Affiliation(s)
- Stephen T. Yeung
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Thomas A. Premeaux
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Li Du
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Toshiro Niki
- Departments of Immunology and Immunopathology, Kagawa University, Kagawa, Japan
| | - Satish K. Pillai
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Kamal M. Khanna
- Department of Microbiology, New York University, New York, NY, United States
- *Correspondence: Lishomwa C. Ndhlovu, ; Kamal M. Khanna,
| | - Lishomwa C. Ndhlovu
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- *Correspondence: Lishomwa C. Ndhlovu, ; Kamal M. Khanna,
| |
Collapse
|
12
|
Bond KH, Sims-Lucas S, Oxburgh L. Targets for Renal Carcinoma Growth Control Identified by Screening FOXD1 Cell Proliferation Pathways. Cancers (Basel) 2022; 14:cancers14163958. [PMID: 36010951 PMCID: PMC9406217 DOI: 10.3390/cancers14163958] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/02/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary FOXD1 regulates the proliferation of clear cell renal cell carcinoma (ccRCC) cells, and ccRCC cells in which FOXD1 has been inactivated do not form tumors efficiently in an animal model. Reproducing growth inhibition in tumor cells by inhibiting FOXD1 pathways presents a possible therapeutic approach for ccRCC and other cancers. We have established an analysis strategy to identify FOXD1-regulated target pathways that may be therapeutically tractable, and compounds that modulate these pathways were selected for testing. Targets in three pathways were identified: FOXM1, PME1, and TMEM167A, which were inhibited by compounds FDI-6, AMZ-30, and silibinin, respectively. The effects of these compounds on the growth of tumor cells from patients cultured in a novel 3D tumor-replica culture environment revealed that FDI-6 and silibinin had strong growth inhibitory effects. This investigation informs new therapeutic targets to control ccRCC tumor growth, and provides a strategy to compare the responsiveness of individual patient tumor replicas to growth-inhibitory compounds. Abstract Clinical association studies suggest that FOXD1 is a determinant of patient outcome in clear cell renal cell carcinoma (ccRCC), and laboratory investigations have defined a role for this transcription factor in controlling the growth of tumors through regulation of the G2/M cell cycle transition. We hypothesized that the identification of pathways downstream of FOXD1 may define candidates for pharmacological modulation to suppress the G2/M transition in ccRCC. We developed an analysis pipeline that utilizes RNA sequencing, transcription factor binding site analysis, and phenotype validation to identify candidate effectors downstream from FOXD1. Compounds that modulate candidate pathways were tested for their ability to cause growth delay at G2/M. Three targets were identified: FOXM1, PME1, and TMEM167A, which were targeted by compounds FDI-6, AMZ-30, and silibinin, respectively. A 3D ccRCC tumor replica model was used to investigate the effects of these compounds on the growth of primary cells from five patients. While silibinin reduced 3D growth in a subset of tumor replicas, FDI-6 reduced growth in all. This study identifies tractable pathways to target G2/M transition and inhibit ccRCC growth, demonstrates the applicability of these strategies across patient tumor replicas, and provides a platform for individualized patient testing of compounds that inhibit tumor growth.
Collapse
Affiliation(s)
- Kyle H. Bond
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
| | - Sunder Sims-Lucas
- Children’s Hospital of Pittsburgh, Rangos Research Building, 4401 Penn Ave, Pittsburgh, PA 15224, USA
| | - Leif Oxburgh
- Rogosin Institute, Room 2-43, 310 East 67th St., New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
13
|
Li X, Huang Z, Zhu L, Yu F, Feng M, Gu A, Jiang J, Wang G, Huang D. Prognostic Model and Nomogram Construction and Validation With an Autophagy-Related Gene Signature in Low-Grade Gliomas. Front Genet 2022; 13:905751. [PMID: 35923699 PMCID: PMC9342864 DOI: 10.3389/fgene.2022.905751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/03/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Autophagy plays a vital role in cancer development. However, the prognostic value of autophagy-related genes (ARGs) in low-grade gliomas (LGG) is unclear. This research aimed to investigate whether ARGs correlated with overall survival (OS) in LGG patients. Methods: RNA-sequencing data were obtained from The Cancer Genome Atlas (TCGA) TARGET GTEx database. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis of ARGs were performed by the “clusterprofile” R package. Cox regression with the wald χ2 test was employed to identify prognostic significant ARGs. Next, the receiver operator characteristic curves were established to evaluate the feasibility of risk score (riskscore=h0(t)exp(∑j=1nCoefj×Xj)) and other clinical risk factors to predict prognosis. A nomogram was constructed. Correlations between clinical features and ARGs were further verified by a t-test or Kruskal–Wallis test. In addition, the correlations between autophagy and immune cells were assessed through the single-sample gene set enrichment analysis (ssGSEA) and tumor immune estimation resource database. Last, the prediction model was verified by LGG data downloaded from the Chinese Glioma Genome Atlas (CGGA) database. Results: Overall, 35 DE-ARGs were identified. Functional enrichment analysis showed that these genes were mainly related to oxidative stress and regulation of autophagy. Nine ARGs (BAX, BIRC5, CFLAR, DIRAS3, GRID2, MAPK9, MYC, PTK6, and TP53) were significantly associated with OS. Age (Hazard ratio (HR) = 1.063, 95% CI: 1.046–1.080), grade (HR = 3.412, 95% CI: 2.164–5.379), histological type (HR = 0.556, 95% CI: 0.346–0.893), and risk score (HR = 1.135, 95% CI: 1.104–1.167) were independent prognostic risk factors (all p < 0.05). In addition, BIRC5, CFLAR, DIRAS3, TP53, and risk scores were found to correlate significantly with age and tumor grade (all p < 0.05). Immune cell enrichment analysis demonstrated that the types of immune cells and their expression levels in the high-risk group were significantly different from those in the low-risk group (all p < 0.05). A prognostic nomogram was constructed to predict 1-, 3-, and 5-year survival, and the prognostic value of sorted ARGs were verified in the CGGA database and clinical samples. Conclusion: Our findings suggest that the 9 DE-ARGs’ risk score model could serve as diagnostic and prognostic biomarkers. The prognostic nomograms could be useful for individualized survival prediction and improved treatment strategies.
Collapse
Affiliation(s)
- Xinrui Li
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Huang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Zhu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Yu
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Minghao Feng
- Department of Thoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Aiqin Gu
- Department of Neurosurgery, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
| | - Jianxin Jiang
- Department of Neurosurgery, Taizhou People’s Hospital Affiliated to Nanjing Medical University, Taizhou, China
- *Correspondence: Jianxin Jiang, ; Guangxue Wang, ; Dongya Huang,
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Jianxin Jiang, ; Guangxue Wang, ; Dongya Huang,
| | - Dongya Huang
- Department of Neurology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Jianxin Jiang, ; Guangxue Wang, ; Dongya Huang,
| |
Collapse
|
14
|
Wu L, Zheng Y, Ruan X, Wu D, Xu P, Liu J, Wu D, Li X. Long-chain noncoding ribonucleic acids affect the survival and prognosis of patients with esophageal adenocarcinoma through the autophagy pathway: construction of a prognostic model. Anticancer Drugs 2022; 33:e590-e603. [PMID: 34338240 PMCID: PMC8670349 DOI: 10.1097/cad.0000000000001189] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/29/2021] [Indexed: 02/05/2023]
Abstract
Autophagy-related long-chain noncoding ribonucleic acids play a vital role in the development of esophageal adenocarcinoma. This study aimed to construct a prognostic model of autophagy-related long-chain noncoding ribonucleic acids and identify potential therapeutical targets for esophageal adenocarcinoma. We downloaded 261 long-chain noncoding RNA transcript samples and clinical data of 87 esophageal adenocarcinoma patients from the Cancer Genome Atlas and 307 autophagy-related genes from www.autophagy.com. We performed Kyoto Encyclopedia of Genes and Genomes and Gene Ontology enrichment analyses and Gene Set Enrichment Analysis to determine risk characteristics and bioinformatics functions of signal transduction pathways. Univariate and multivariate Cox regression analyses were used to determine the correlation between autophagy-related long-chain noncoding ribonucleic acids and independent risk factors. The receiver operating characteristic analysis was used to evaluate the feasibility of the prognostic model. Finally, we performed survival analysis, risk analysis and independent prognostic analysis to verify the prognostic model of esophageal adenocarcinoma. We identified 22 autophagic long-chain noncoding ribonucleic acids that were highly correlated with the overall survival of esophageal adenocarcinoma patients. The areas under the receiver operating characteristic curve (0.941) and the calibration curve were significantly similar. Moreover, univariate and multivariate Cox regression analyses indicated that autophagy-related long-chain noncoding ribonucleic acids were independent predictors of esophageal adenocarcinoma. We found that autophagy-related long-chain noncoding ribonucleic acids might affect tumor development and prognosis in esophageal adenocarcinoma patients. The findings indicate that the prognostic model of esophageal adenocarcinoma has potential therapeutic applications in patients with esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Liusheng Wu
- Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, Guangdong
- Department of graduate school, Anhui Medical University, Hefei, Anhui
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| | - Yuzhen Zheng
- Department of Thoracic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou
| | - Xin Ruan
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
- Shantou University Medical College, Shantou, Guangdong, China
| | - Dingwang Wu
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| | - Pengcheng Xu
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| | - Jixian Liu
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| | - Da Wu
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| | - Xiaoqiang Li
- Peking University Shenzhen Hospital, Clinical College of Anhui Medical University, Shenzhen, Guangdong
- Department of thoracic surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong
| |
Collapse
|
15
|
Zargar Balajam N, Shabani M, Aghaei M. Galectin-9 inhibits cell proliferation and induces apoptosis in Jurkat and KE-37 acute lymphoblastic leukemia cell lines via caspase-3 activation. Res Pharm Sci 2021; 16:612-622. [PMID: 34760009 PMCID: PMC8562407 DOI: 10.4103/1735-5362.327507] [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: 10/28/2021] [Revised: 04/06/2021] [Accepted: 09/12/2021] [Indexed: 11/27/2022] Open
Abstract
Background and purpose: Acute lymphoblastic leukemia (ALL) is a type of cancer of blood and bone marrow characterized by abnormal proliferation of lymphoid progenitor cells. Galectin-9 is a tandem-repeat type galectin expressed in various tumor cells. It seems that the connection between galectin-9 and T cell immunoglobulin mucin-3 receptor acts as a negative regulator of cancer cells proliferation. Experimental approach: In this research, the effects of galectin-9 were investigated using MTS cell proliferation colorimetric, colony-forming, annexin V-FITC/PI, and caspase-3 assays in the Jurkat and KE-37 cell lines of ALL. Furthermore, the western blotting technique was used to evaluate the levels of apoptotic proteins such as Bax and Bcl-2 in these cell lines. Findings/Results: Our results indicated that galectin-9 can considerably reduce the cell growth and colony formation ability of both Jurkat and KE-37 cell lines in a concentration-dependent manner. Besides, galectin-9 induced apoptosis in a concentration-dependent manner in ALL cells by a mechanism associated with Bax/Bcl-2 expression and activation of the caspase-3 activation. Conclusion and implications: Galectin-9 inhibited the growth and proliferation of cell lines with increased programmed cell death, therefore it can be considered as a potential factor in the progression of ALL therapeutics that needs more research in this context.
Collapse
Affiliation(s)
- Narges Zargar Balajam
- Department of Clinical Biochemistry and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mahdi Shabani
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, I.R. Iran
| | - Mahmoud Aghaei
- Department of Clinical Biochemistry and Isfahan Pharmaceutical Sciences Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
16
|
Kremsreiter SM, Kroell ASH, Weinberger K, Boehm H. Glycan-Lectin Interactions in Cancer and Viral Infections and How to Disrupt Them. Int J Mol Sci 2021; 22:10577. [PMID: 34638920 PMCID: PMC8508825 DOI: 10.3390/ijms221910577] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glycan-lectin interactions play an essential role in different cellular processes. One of their main functions is involvement in the immune response to pathogens or inflammation. However, cancer cells and viruses have adapted to avail themselves of these interactions. By displaying specific glycosylation structures, they are able to bind to lectins, thus promoting pathogenesis. While glycan-lectin interactions promote tumor progression, metastasis, and/or chemoresistance in cancer, in viral infections they are important for viral entry, release, and/or immune escape. For several years now, a growing number of investigations have been devoted to clarifying the role of glycan-lectin interactions in cancer and viral infections. Various overviews have already summarized and highlighted their findings. In this review, we consider the interactions of the lectins MGL, DC-SIGN, selectins, and galectins in both cancer and viral infections together. A possible transfer of ways to target and disrupt them might lead to new therapeutic approaches in different pathological backgrounds.
Collapse
Affiliation(s)
- Stefanie Maria Kremsreiter
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Ann-Sophie Helene Kroell
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Katharina Weinberger
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Ruprecht Karls University Heidelberg, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany; (S.M.K.); (A.-S.H.K.); (K.W.)
| | - Heike Boehm
- Max-Planck-Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
| |
Collapse
|
17
|
Zhu L, Dong L, Feng M, Yang F, Jiang W, Huang Z, Liu F, Wang L, Wang G, Li Q. Profiles of autophagy-related genes in esophageal adenocarcinoma. BMC Cancer 2020; 20:943. [PMID: 32998713 PMCID: PMC7528598 DOI: 10.1186/s12885-020-07416-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 09/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background Several studies have demonstrated autophagy was involved in the process of esophageal adenocarcinoma (EAC). The aim of this study was to explore autophagy-related genes (ARGs) correlated with overall survival (OS) in EAC patients. Methods Expressions of ARGs in EAC and normal samples were downloaded from TCGA database. GO and KEGG enrichment analyses were used to investigate the ARGs bioinformatics functions. Univariate and multivariate cox regressions were performed to identify prognostic ARGs and the independent risk factors. ROC curve was established to evaluate the feasibility to predict the prognosis. Finally, the correlations between ARGs and clinical features were further explored. In addition, significantly different ARGs were verified in EAC specimens and normal esophageal mucosal tissues. Results Thirty significantly different ARGs were selected from EAC and normal tissues. Functional enrichments showed these ARGs were mainly related apoptosis. Multivariate cox regression analyses demonstrated eight ARGs were significantly associated with OS. Among these eight genes, BECN1 (HR = 0.321, P = 0.046), DAPK1 (HR = 0.636, P = 0.025) and CAPN1 (HR = 0.395, P = 0.004) played protective roles in survival. Gender (HR = 0.225, P = 0.032), stage (HR = 5.841, P = 0.008) and risk score (HR = 1.131, P < 0.001) were independent prognostic risk factors. ROC curves showed better efficacy to predict survival using the risk score. Additionally, we found BECN1, DAPK1, VAMP7 and SIRT1 genes were correlated significantly with survival status, gender, primary tumor and tumor stage (all P < 0.05). The experimental results confirmed the BIRC5 was overexpressed and the ITPR1, PRKN were downregulated in the EAC tissues compared with the normal esophageal mucosal tissues (all P < 0.05). Conclusion Our findings suggested that autophagy was involved in the process of EAC. Several ARGs probably could serve as diagnostic and prognostic biomarkers and may help facilitate therapeutic targets in EAC patients.
Collapse
Affiliation(s)
- Lei Zhu
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Lin Dong
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Minghao Feng
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Fugui Yang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Wenhao Jiang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Zhiyuan Huang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Fabing Liu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, 200080, China
| | - Lingwei Wang
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China
| | - Guangxue Wang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| | - Qinchuan Li
- Department of Thoracic Surgery, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China. .,Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, 150 Jimo Road, Shanghai, 200120, China.
| |
Collapse
|
18
|
Shete A, Bichare S, Pujari V, Virkar R, Thakar M, Ghate M, Patil S, Vyakarnam A, Gangakhedkar R, Bai G, Niki T, Hattori T. Elevated Levels of Galectin-9 but Not Osteopontin in HIV and Tuberculosis Infections Indicate Their Roles in Detecting MTB Infection in HIV Infected Individuals. Front Microbiol 2020; 11:1685. [PMID: 32765475 PMCID: PMC7380070 DOI: 10.3389/fmicb.2020.01685] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/29/2020] [Indexed: 12/12/2022] Open
Abstract
Galectin-9 (Gal-9) and osteopontin (OPN) play immunomodulatory roles in tuberculosis and HIV infections. Evaluation of their levels as well as their interplay with different pro-inflammatory cytokines is critical to understand their role in immunopathogenesis of HIV/tuberculosis co-infection considering the complexity of the disease. Plasma levels of these proteins were measured by ELISAs in HIV-negative individuals with pulmonary (n = 21), extrapulmonary (n = 33), and latent tuberculosis (n = 22) and in HIV infected patients with pulmonary (n = 14), latent tuberculosis (n = 17), and without tuberculosis (n = 41). Levels of pro-inflammatory cytokines were estimated by Luminex assay. Receiver operated characteristic curve analysis was performed to evaluate discriminatory roles of these proteins. Spearman's correlation analysis was performed with the markers of HIV and tuberculosis disease progression to evaluate their immunopathogenic roles. Gal-9 and OPN levels were higher in HIV uninfected patients with active tuberculosis than with latent tuberculosis. Gal-9 but not OPN levels were higher in HIV infected patients with active tuberculosis than with latent tuberculosis. Area under curve for Galectin-9 was >0.9 in HIV/tuberculosis co-infection and extrapulmonary tuberculosis. OPN and IL-6 levels were higher in patients with severe chest X-ray grade indicating its association with severity of the disease and positively correlated with each other. Stronger positive and negative correlations of Gal-9 levels, respectively, with viral loads and CD4 cell counts in HIV infected patients were observed than OPN levels indicating their association with HIV disease progression. Thus, significantly elevated Gal-9 levels were reported for the first time in HIV/tuberculosis co-infection and extrapulmonary tuberculosis in our study than single infections with HIV and tuberculosis. The study indicated a need for further evaluation of monitoring role of Gal-9 for detection of developing tuberculosis in HIV infected individuals. The findings also indicated differential roles of Gal-9 and OPN in the pathogenesis of tuberculosis and HIV infections.
Collapse
Affiliation(s)
| | | | | | - Rashmi Virkar
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, India
| | | | | | - Sandip Patil
- ICMR-National AIDS Research Institute, Pune, India
| | | | | | - Gaowa Bai
- Department of Health Science and Social Welfare, Kibi International University, Takahashi, Japan
| | - Toshiro Niki
- Department of Immunology, Faculty of Medicine, Kagawa University, Kita-gun, Japan
| | - Toshio Hattori
- Department of Health Science and Social Welfare, Kibi International University, Takahashi, Japan
| |
Collapse
|
19
|
Esophageal Cancer Development: Crucial Clues Arising from the Extracellular Matrix. Cells 2020; 9:cells9020455. [PMID: 32079295 PMCID: PMC7072790 DOI: 10.3390/cells9020455] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
In the last years, the extracellular matrix (ECM) has been reported as playing a relevant role in esophageal cancer (EC) development, with this compartment being related to several aspects of EC genesis and progression. This sounds very interesting due to the complexity of this highly incident and lethal tumor, which takes the sixth position in mortality among all tumor types worldwide. The well-established increase in ECM stiffness, which is able to trigger mechanotransduction signaling, is capable of regulating several malignant behaviors by converting alteration in ECM mechanics into cytoplasmatic biochemical signals. In this sense, it has been shown that some molecules play a key role in these events, particularly the different collagen isoforms, as well as enzymes related to its turnover, such as lysyl oxidase (LOX) and matrix metalloproteinases (MMPs). In fact, MMPs are not only involved in ECM stiffness, but also in other events related to ECM homeostasis, which includes ECM remodeling. Therefore, the crucial role of distinct MMPs isoform has already been reported, especially MMP-2, -3, -7, and -9, along EC development, thus strongly associating these proteins with the control of important cellular events during tumor progression, particularly in the process of invasion during metastasis establishment. In addition, by distinct mechanisms, a vast diversity of glycoproteins and proteoglycans, such as laminin, fibronectin, tenascin C, galectin, dermatan sulfate, and hyaluronic acid exert remarkable effects in esophageal malignant cells due to the activation of oncogenic signaling pathways mainly involved in cytoskeleton alterations during adhesion and migration processes. Finally, the wide spectrum of interactions potentially mediated by ECM may represent a singular intervention scenario in esophageal carcinogenesis natural history and, due to the scarce knowledge on the cellular and molecular mechanisms involved in EC development, the growing body of evidence on ECM’s role along esophageal carcinogenesis might provide a solid base to improve its management in the future.
Collapse
|
20
|
Wiersma VR. Lectins as modulators of autophagy in cancer immunotherapy. AUTOPHAGY IN IMMUNE RESPONSE: IMPACT ON CANCER IMMUNOTHERAPY 2020:53-74. [DOI: 10.1016/b978-0-12-819609-0.00004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Possible therapeutic applicability of galectin-9 in cutaneous T-cell lymphoma. J Dermatol Sci 2019; 96:134-142. [PMID: 31787505 DOI: 10.1016/j.jdermsci.2019.09.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/26/2019] [Accepted: 09/10/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Galectin-9, a member of the galectin family, can promote tumor growth through inducing apoptosis in anti-tumor immune cells via T cell immunoglobulin and mucin domain 3 (TIM-3). On the other hand, galectin-9 also induces tumor cell apoptosis in many malignancies and thought to have potential as an anti-cancer agent. OBJECTIVE To examine the expression and therapeutic applicability of galectin-9 in cutaneous T-cell lymphoma (CTCL). METHODS Galectin-9 expression in lesional skin and sera was measured using CTCL samples. The effect of galectin-9 on CTCL cell lines was investigated in vitro. We also examined effect of galectin-9 on tumor growth of CTCL cells in immune-deficient mice. Moreover, we examined the efficacy of galectin-9, anti-TIM-3 blocking antibody, or their combination on tumor growth of EL-4 cells in wild-type mice. RESULTS Galectin-9 was expressed on tumor cells in lesional skin of CTCL and the expression levels were associated with decreased CD8+ T-cell infiltration. Serum galectin-9 levels were correlated with disease severity markers. High-dose galectin-9 induced cell death of CTCL cell lines through activation of caspase-3 and caspase-9, independently of TIM-3. High-dose galectin-9 suppressed the growth of CTCL cells and EL-4 cells in vivo. Furthermore, additional anti-TIM-3 blocking antibody administration to galectin-9 achieved greater inhibition of tumor growth compared to single administration. CONCLUSION Galectin-9 expression on tumor cells may be associated with CTCL progression through attenuating anti-tumor immunity. On the other hand, exogenous high-dose galectin-9 administration can be a therapeutic strategy for CTCL and anti-TIM-3 blocking antibody can augment the efficacy of galectin-9.
Collapse
|
22
|
Chiyo T, Fujita K, Iwama H, Fujihara S, Tadokoro T, Ohura K, Matsui T, Goda Y, Kobayashi N, Nishiyama N, Yachida T, Morishita A, Kobara H, Mori H, Niki T, Hirashima M, Himoto T, Masaki T. Galectin-9 Induces Mitochondria-Mediated Apoptosis of Esophageal Cancer In Vitro and In Vivo in a Xenograft Mouse Model. Int J Mol Sci 2019; 20:ijms20112634. [PMID: 31146370 PMCID: PMC6600680 DOI: 10.3390/ijms20112634] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 12/30/2022] Open
Abstract
Galectin-9 (Gal-9) enhances tumor immunity mediated by T cells, macrophages, and dendritic cells. Its expression level in various cancers correlates with prognosis. Furthermore, Gal-9 directly induces apoptosis in various cancers; however, its mechanism of action and bioactivity has not been clarified. We evaluated Gal-9 antitumor effect against esophageal squamous cell carcinoma (ESCC) to analyze the dynamics of apoptosis-related molecules, elucidate its mechanism of action, and identify relevant changes in miRNA expressions. KYSE-150 and KYSE-180 cells were treated with Gal-9 and their proliferation was evaluated. Gal-9 inhibited cell proliferation in a concentration-dependent manner. The xenograft mouse model established with KYSE-150 cells was administered with Gal-9 and significant suppression in the tumor growth observed. Gal-9 treatment of KYSE-150 cells increased the number of Annexin V-positive cells, activation of caspase-3, and collapse of mitochondrial potential, indicating apoptosis induction. c-Jun NH2-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38) phosphorylation were activated and could be involved in apoptosis. Therefore, Gal-9 induces mitochondria-mediated apoptosis of ESCC and inhibits cell proliferation in vitro and in vivo with JNK and p38 activation.
Collapse
Affiliation(s)
- Taiga Chiyo
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Koji Fujita
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Hisakazu Iwama
- Life Science Research Center, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Shintaro Fujihara
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Tomoko Tadokoro
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Kyoko Ohura
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Takanori Matsui
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Yasuhiro Goda
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Nobuya Kobayashi
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Noriko Nishiyama
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Tatsuo Yachida
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Hirohito Mori
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Toshiro Niki
- Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Mitsuomi Hirashima
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| | - Takashi Himoto
- Department of Medical Technology, Kagawa Prefectural University of Health Sciences, 281-1, Hara, Mure-Cho, Takamatsu, Kagawa 761-0123, Japan.
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, 1750-1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan.
| |
Collapse
|
23
|
Pan P, Dombkowski AA, Wang LS, Stoner GD. A nutrigenetic approach for investigating the chemopreventive effects of black raspberries during the development of preneoplastic esophagi in rats. JOURNAL OF BERRY RESEARCH 2018; 8:263-274. [PMID: 30613310 PMCID: PMC6319902 DOI: 10.3233/jbr-180346] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
BACKGROUND Large epidemiological studies have shown that diets high in fruits reduce the risk of esophageal squamous cell carcinoma (ESCC). OBJECTIVE The current study investigated the effects of black raspberries (BRBs) on gene expression during the development of preneoplastic esophagi in rats. METHODS Using a post-initiation protocol, F344 rats were injected with N-nitrosomethylbenzylamine (NMBA) and then fed either a control diet or 5% BRBs. At weeks 9, 15, and 35, we euthanized subgroups of the rats and collected preneoplastic esophagi to isolate RNA samples for DNA microarray. RESULTS Along the development of NMBA-induced preneoplastic esophagi, NMBA injections led to differential expression of 1181 genes comparing to control rats, and dietary BRBs modulated 428 genes in esophagi from NMBA-treated rats. There are 137 common genes between 1181 and 428 gene sets, and BRBs significantly reversed the expression of 133 genes. These genes are associated with multiple gene oncology functions. BRBs induced an 8.8-fold gene enrichment on the pathway of inflammatory response and regulated 10 genes involved in this pathway. Among them, BRBs significantly reversed the expression of pro-inflammatory cytokines, such as CCL2, S100A8, and IL19. CONCLUSIONS BRBs exhibit strong anti-inflammatory effects against NMBA-induced rat esophageal tumorigenesis.
Collapse
Affiliation(s)
- Pan Pan
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Alan A. Dombkowski
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Li-Shu Wang
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Gary D. Stoner
- Department of Medicine, Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
24
|
Li X, Zhu X, Xu C, Wu J. 2-Methyl 2-butanol suppresses human retinoblastoma cells through cell cycle arrest and autophagy. ACTA ACUST UNITED AC 2018; 51:e6889. [PMID: 29561956 PMCID: PMC5875906 DOI: 10.1590/1414-431x20176889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/28/2017] [Indexed: 01/01/2023]
Abstract
2-Methyl-2-butanol (MBT) is a chemical compound from the group of alcohols more specifically pentanols, which has shown an excellent anti-cancer activity in our previous study. However, its mechanism of action remains unclear. The present study was designed to investigate the anti-cancer effect of MBT on human retinoblastoma cells. The results showed that the use of MBT leads to HXO-RB44 cell death but is cytotoxic to normal cells at higher concentrations. It showed a dose- as well as a time-dependent inhibition of HXO-RB44 cells. P27 is a cell cycle inhibitory protein, which plays an important role in cell cycle regulation whereas cyclin-B1 is a regulatory protein involved in mitosis. MBT increased the cell cycle arrest in a dose-dependent manner by augmenting p27 and reducing cyclin B1 expression. Moreover, it also accelerated apoptosis, increased light chain-3 (LC-3) conversion in a dose-dependent manner, and helped to debulk cancerous cells. LC3 is a soluble protein, which helps to engulf cytoplasmic components, including cytosolic proteins and organelles during autophagy from autophagosomes. In order to verify the effect of MBT, bafilomycin A1, an autophagy inhibitor, was used to block the MTB-induced apoptosis and necrosis. Additionally, a specific Akt agonist, SC-79, reversed the MBT-induced cell cycle arrest and autophagy. Thus, from the present study, it was concluded that MBT induced cell cycle arrest, apoptosis and autophagy through the PI3K/Akt pathway in HXO-RB44 cells.
Collapse
Affiliation(s)
- Xiangyun Li
- Department of Fundus, Wuhan Aier Eye Hospital, Wuhan, Hubei Province, China
| | - Xiangxiang Zhu
- Ophthalmology Center, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Chong Xu
- Department of Fundus, Wuhan Aier Eye Hospital, Wuhan, Hubei Province, China
| | - Jianhua Wu
- Department of Fundus, Wuhan Aier Eye Hospital, Wuhan, Hubei Province, China
| |
Collapse
|