1
|
Zhang L, Chen W, Li X, Wang G, Xing F, Zhu X. Galectin-1 overexpression induces normal fibroblasts translate into cancer-associated fibroblasts and attenuates the sensitivity of anlotinib in lung cancer. Cell Adh Migr 2024; 18:1-11. [PMID: 38557441 PMCID: PMC10986763 DOI: 10.1080/19336918.2024.2335881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
We aimed to investigate galectin-1 overexpression induces normal fibroblasts (NFs) translates into cancer-associated fibroblasts (CAFs). Galectin-1 overexpression was conducted in Human embryonic lung fibroblasts (HFL1) cell. The motilities of H1299 and A549 cells were measured. Human umbilical vein endothelial cell (HUVEC) proliferation and tube formation ability were assessed. Tumor volume and tumor weight was recorded. Cells motilities were increased, while apoptosis rates were decreased after CMs co-cultured. B-cell lymphoma-2 (Bcl-2) expression level was increased, while Bcl2-associatedX (Bax) and cleaved-caspase3 decreased. CMs treatment enhanced HUVEC proliferation and tube formation. Tumor volume and weight in CMs treated mice were increased, and the sensitivity of anlotinib in co-cultured cells was decreased. Our results revealed that galectin-1 overexpression induced NFs translated into CAFs.
Collapse
Affiliation(s)
- Lei Zhang
- 0Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Wenbang Chen
- 0Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaojun Li
- 0Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Gengming Wang
- Department of Radiotherapy, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Fubao Xing
- 0Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| | - Xiao Zhu
- 0Department of Thoracic Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
2
|
Troncoso MF, Elola MT, Blidner AG, Sarrias L, Espelt MV, Rabinovich GA. The universe of galectin-binding partners and their functions in health and disease. J Biol Chem 2023; 299:105400. [PMID: 37898403 PMCID: PMC10696404 DOI: 10.1016/j.jbc.2023.105400] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/30/2023] Open
Abstract
Galectins, a family of evolutionarily conserved glycan-binding proteins, play key roles in diverse biological processes including tissue repair, adipogenesis, immune cell homeostasis, angiogenesis, and pathogen recognition. Dysregulation of galectins and their ligands has been observed in a wide range of pathologic conditions including cancer, autoimmune inflammation, infection, fibrosis, and metabolic disorders. Through protein-glycan or protein-protein interactions, these endogenous lectins can shape the initiation, perpetuation, and resolution of these processes, suggesting their potential roles in disease monitoring and treatment. However, despite considerable progress, a full understanding of the biology and therapeutic potential of galectins has not been reached due to their diversity, multiplicity of cell targets, and receptor promiscuity. In this article, we discuss the multiple galectin-binding partners present in different cell types, focusing on their contributions to selected physiologic and pathologic settings. Understanding the molecular bases of galectin-ligand interactions, particularly their glycan-dependency, the biochemical nature of selected receptors, and underlying signaling events, might contribute to designing rational therapeutic strategies to control a broad range of pathologic conditions.
Collapse
Affiliation(s)
- María F Troncoso
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María T Elola
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Ada G Blidner
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Luciana Sarrias
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María V Espelt
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina; Instituto de Química y Fisicoquímica Biológicas (IQUIFIB) Prof Alejandro C. Paladini, CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
3
|
ESCCAL-1 promotes cell-cycle progression by interacting with and stabilizing galectin-1 in esophageal squamous cell carcinoma. NPJ Precis Oncol 2022; 6:12. [PMID: 35233069 PMCID: PMC8888636 DOI: 10.1038/s41698-022-00255-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 02/03/2022] [Indexed: 11/20/2022] Open
Abstract
Long non-coding RNAs (LncRNAs) play important roles in the development of human esophageal squamous cell carcinoma (ESCC). Our previous studies have shown that knockdown of LncRNA ESCCAL-1 expression inhibits the growth of ESCC cells, but the mechanisms remain largely unknown. In this study, we show that over-expression of ESCCAL-1 promotes ESCC cell proliferation and cell-cycle progression by blocking ubiquitin-mediated degradation of an oncoprotein galectin-1 (Gal-1). Multiple LncRNA expression datasets as well as our own data together reveal that ESCCAL-1 is evidently up-regulated in ESCC tissues and exhibits promising diagnostic value. Over-expression of ESCCAL-1 augmented ESCC cell proliferation and cell-cycle progression, whereas down-regulation of ESCCAL-1 resulted in the opposite effects. Mechanistically, LncRNA ESCCAL-1 directly binds to Gal-1 and positively regulates its protein level without affecting its mRNA level. Up-regulation of Gal-1 facilitated ESCC cell proliferation and cell-cycle progress. Knockdown of Gal-1 mitigated the effects of ESCCAL-1-mediated high cellular proliferation, NF-κB signaling activation and tumorigenicity of ESCC cells. Thus, our findings provide novel insight into the mechanism by which ESCCAL-1 facilitates ESCC tumorigenesis and cell-cycle progression by interacting with and stabilizing Gal-1 protein, suggesting a potential therapeutic target for ESCC.
Collapse
|
4
|
Carabias P, Espelt MV, Bacigalupo ML, Rojas P, Sarrias L, Rubin A, Saffioti NA, Elola MT, Rossi JP, Wolfenstein-Todel C, Rabinovich GA, Troncoso MF. Galectin-1 confers resistance to doxorubicin in hepatocellular carcinoma cells through modulation of P-glycoprotein expression. Cell Death Dis 2022; 13:79. [PMID: 35075112 PMCID: PMC8786848 DOI: 10.1038/s41419-022-04520-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/15/2022]
Abstract
Galectin-1 (GAL1), a β-galactoside-binding protein abundantly expressed in the tumor microenvironment, has emerged as a key mechanism of chemoresistance developed by different tumors. Although increased expression of GAL1 is a hallmark of hepatocellular carcinoma (HCC) progression, aggressiveness and metastasis, limited information is available on the role of this endogenous lectin in HCC resistance to chemotherapy. Moreover, the precise mechanisms underlying this effect are uncertain. HCC has evolved different mechanisms of resistance to chemotherapy including those involving the P-glycoprotein (P-gp), an ATP-dependent drug efflux pump, which controls intracellular drug concentration. Here, we investigated the molecular mechanism underlying GAL1-mediated chemoresistance in HCC cells, particularly the involvement of P-gp in this effect. Our results show that GAL1 protected HepG2 cells from doxorubicin (DOX)- and sorafenib-induced cell death in vitro. Accordingly, GAL1-overexpressing HepG2 cells generated DOX-resistant tumors in vivo. High expression of GAL1 in HepG2 cells reduced intracellular accumulation of DOX likely by increasing P-gp protein expression rather than altering its membrane localization. GAL1-mediated increase of P-gp expression involved activation of the phosphatidylinositol-3 kinase (PI3K) signaling pathway. Moreover, 'loss-of-function' experiments revealed that P-gp mediates GAL1-driven resistance to DOX, but not to sorafenib, in HepG2 cells. Conversely, in PLC/PRF/5 cells, P-gp protein expression was undetectable and GAL1 did not control resistance to DOX or sorafenib, supporting the critical role of P-gp in mediating GAL1 effects. Collectively, our findings suggest that GAL1 confers chemoresistance in HCC through mechanisms involving modulation of P-gp, thus emphasizing the role of this lectin as a potential therapeutic target in HCC.
Collapse
Grants
- PICT-2014-3216 Ministerio de Ciencia, Tecnología e Innovación Productiva (Ministry of Science, Technology and Productive Innovation, Argentina)
- PICT V 2014-3687 Ministerio de Ciencia, Tecnología e Innovación Productiva (Ministry of Science, Technology and Productive Innovation, Argentina)
- PICT-2016-1139 Ministerio de Ciencia, Tecnología e Innovación Productiva (Ministry of Science, Technology and Productive Innovation, Argentina)
- 20020150100005BA Universidad de Buenos Aires (University of Buenos Aires)
- PIP-11220150100647 Consejo Nacional de Investigaciones Científicas y Técnicas (National Scientific and Technical Research Council)
- Sales, Bunge & Born and Lounsbery Foundations. Donations from the Ferioli, Ostry and Caraballo families.
Collapse
Affiliation(s)
- Pablo Carabias
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - María V Espelt
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - María L Bacigalupo
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Paola Rojas
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Luciana Sarrias
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Ayelén Rubin
- Laboratorio de Carcinogénesis Hormonal, Instituto de Biología y Medicina Experimental, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Nicolás A Saffioti
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - María T Elola
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Juan P Rossi
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Carlota Wolfenstein-Todel
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Buenos Aires, Argentina
- Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F Troncoso
- Universidad de Buenos Aires, Consejo Nacional de lnvestigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Buenos Aires, Argentina.
| |
Collapse
|
5
|
Zheng Y, Wang S, Cai J, Ke A, Fan J. The progress of immune checkpoint therapy in primary liver cancer. Biochim Biophys Acta Rev Cancer 2021; 1876:188638. [PMID: 34688805 DOI: 10.1016/j.bbcan.2021.188638] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 12/17/2022]
Abstract
After years of in-depth research on immune checkpoints, therapeutic reversal of immune-exhaustion with immune checkpoint inhibitors (ICPIs) has been shown to be effective in primary liver cancer, including hepatocellular carcinoma and intrahepatic cholangiocarcinoma. The clinical development of novel ICPIs continues at a rapid pace, with more than 200 clinical trials of immunotherapeutic agents registered as of July 2021 for the treatment of liver cancer. In this review, we discussed the immune tolerance mechanism of liver cancer and the biological basis of immune checkpoints, focusing on the current status of ICPIs' development and clinical application. In addition, ICPIs combined with local resection, radiofrequency ablation, chemoembolization, and other molecular targeted drug therapies have shown better efficacy. Combined therapy based on multidisciplinary cooperation is the future direction of treatment in liver cancer.
Collapse
Affiliation(s)
- Yimin Zheng
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, PR China; Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Siwei Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, PR China
| | - Jiabin Cai
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Aiwu Ke
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, PR China.
| | - Jia Fan
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai 200032, PR China; Department of Liver Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| |
Collapse
|
6
|
Galectins in Cancer and the Microenvironment: Functional Roles, Therapeutic Developments, and Perspectives. Biomedicines 2021; 9:biomedicines9091159. [PMID: 34572346 PMCID: PMC8465754 DOI: 10.3390/biomedicines9091159] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/15/2022] Open
Abstract
Changes in cell growth and metabolism are affected by the surrounding environmental factors to adapt to the cell’s most appropriate growth model. However, abnormal cell metabolism is correlated with the occurrence of many diseases and is accompanied by changes in galectin (Gal) performance. Gals were found to be some of the master regulators of cell–cell interactions that reconstruct the microenvironment, and disordered expression of Gals is associated with multiple human metabolic-related diseases including cancer development. Cancer cells can interact with surrounding cells through Gals to create more suitable conditions that promote cancer cell aggressiveness. In this review, we organize the current understanding of Gals in a systematic way to dissect Gals’ effect on human disease, including how Gals’ dysregulated expression affects the tumor microenvironment’s metabolism and elucidating the mechanisms involved in Gal-mediated diseases. This information may shed light on a more precise understanding of how Gals regulate cell biology and facilitate the development of more effective therapeutic strategies for cancer treatment by targeting the Gal family.
Collapse
|
7
|
Chu H, Wu C, Zhao Q, Sun R, Yang K, Zhao B, Liu Y, Liang Z, Zhong S, Zhang L, Zhang Y. Quantitative proteomics identifies FOLR1 to drive sorafenib resistance via activating autophagy in hepatocellular carcinoma cells. Carcinogenesis 2021; 42:753-761. [PMID: 33677528 DOI: 10.1093/carcin/bgab019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/03/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
Sorafenib is commonly used to treat advanced human hepatocellular carcinoma (HCC). However, clinical efficacy has been limited by drug resistance. In this study, we used label-free quantitative proteomic analysis to systematically investigate the underlying mechanisms of sorafenib resistance in HCC cells. A total of 1709 proteins were confidently quantified. Among them, 89 were differentially expressed and highly enriched in the processes of cell-cell adhesion, negative regulation of apoptosis, response to drug and metabolic processes involving in sorafenib resistance. Notably, folate receptor α (FOLR1) was found to be significantly upregulated in resistant HCC cells. In addition, in vitro studies showed that overexpression of FOLR1 decreased the sensitivity of HCC cells to sorafenib, whereas siRNA-directed knockdown of FOLR1 increased the sensitivity of HCC cells to sorafenib. Immunoprecipitation-mass spectrometry analysis suggested a strong link between FOLR1 and autophagy-related proteins. Further biological experiments found that FOLR1-related sorafenib resistance was accompanied by the activation of autophagy, whereas inhibition of autophagy significantly reduced FOLR1-induced cell resistance. These results suggest the driving role of FOLR1 in HCC resistance to sorafenib, which may be exerted through FOLR1-induced autophagy. Therefore, this study may provide new insights into understanding the mechanism of sorafenib resistance.
Collapse
Affiliation(s)
- Hongwei Chu
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Changqing Wu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China.,Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Qun Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Rui Sun
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Kuo Yang
- BNRIST/Department of Automation, Tsinghua University, Beijing, China
| | - Baofeng Zhao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Yang Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China.,Innovative Research Center for Integrated Cancer Omics, the Second Hospital Affiliated to China Medical University, Shenyang, China
| | - Zhen Liang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Shijun Zhong
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| | - Yukui Zhang
- Zhang Dayu School of Chemistry, Dalian University of Technology, Dalian, China.,CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R. & A. Center, Dalian Institute of Chemical Physics, Chinese Academy of Science, Dalian, China
| |
Collapse
|
8
|
Setayesh T, Colquhoun SD, Wan YJY. Overexpression of Galectin-1 and Galectin-3 in hepatocellular carcinoma. LIVER RESEARCH 2020; 4:173-179. [PMID: 34567824 PMCID: PMC8460053 DOI: 10.1016/j.livres.2020.11.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Galectins (Gals) are evolutionarily conserved proteins that bind to β-galactoside containing glycans. Abnormal expression of Gals is associated with the development, progression, and metastasis of different types of cancer. Among the 11 Gals identified in humans, the roles of Gal-1 and Gal-3 have been extensively investigated in various tumors. Here, we summarize the roles of overly expressed Gal-1 and Gal-3 in the pathogenesis of hepatocellular carcinoma (HCC). The overexpression of Gal-1 and Gal-3 correlates with tumor growth, HCC cell migration and invasion, tumor aggressiveness, metastasis, and poor prognosis. A potentially promising future treatment strategy for HCC may include the combination of immunotherapy with Gal-1 inhibition. Additional research is warranted to investigate targeting Gal-1 and Gal-3 for HCC treatment.
Collapse
Affiliation(s)
- Tahereh Setayesh
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA
| | | | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA,Corresponding author. Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA, USA. (Y.-J.Y. Wan)
| |
Collapse
|
9
|
Sun MJ, Cao ZQ, Leng P. The roles of galectins in hepatic diseases. J Mol Histol 2020; 51:473-484. [PMID: 32734557 DOI: 10.1007/s10735-020-09898-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 07/14/2020] [Indexed: 12/24/2022]
Abstract
Hepatic diseases include all diseases that occur in the liver, including hepatitis, cirrhosis, hepatocellular carcinoma, etc. Hepatic diseases worldwide are characterized by high incidences of digestive system diseases, which present with subtle symptoms, are difficult to treat and have high mortality. Galectins are β-galactoside-binding proteins that have been found to be aberrantly expressed during hepatic disease progression. An increasing number of studies have shown that abnormal expression of galectins is extensively involved in hepatic diseases, such as hepatocellular carcinoma (HCC), liver cirrhosis, hepatitis and liver fibrosis. Galectins function as intracellular and extracellular hepatic disease regulators mainly through the binding of their carbohydrate recognition domain to glycoconjugates expressed in hepatocytes. In this review, we summarize current research on the various roles of galectins in cirrhosis, hepatitis, liver fibrosis and HCC, which may provide a preliminary theoretical basis for the exploration of new targets for the treatment of hepatic diseases.
Collapse
Affiliation(s)
- Mei-Juan Sun
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, No. 16 Jiang Su Road, Qingdao, 266003, People's Republic of China
| | - Zhan-Qi Cao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, No. 16 Jiang Su Road, Qingdao, 266003, People's Republic of China
| | - Ping Leng
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, No. 16 Jiang Su Road, Qingdao, 266003, People's Republic of China.
| |
Collapse
|
10
|
Poté N, Cros J, Laouirem S, Raffenne J, Negrão M, Albuquerque M, Bedossa P, Godinho Ferreira M, Ait Si Ali S, Fior R, Paradis V. The histone acetyltransferase hMOF promotes vascular invasion in hepatocellular carcinoma. Liver Int 2020; 40:956-967. [PMID: 31943753 DOI: 10.1111/liv.14381] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 12/27/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Vascular invasion is a major prognostic factor in hepatocellular carcinoma (HCC). We previously identified histone H4 acetylated at lysine 16 (H4K16ac), a histone modification involved in transcription activation, as a biomarker of microvascular invasion (mVI) in HCC. This study aimed to investigate the role of hMOF, the histone acetyltransferase responsible for H4K16 acetylation, in the process of vascular invasion in HCC. METHODS hMOF expression was assessed by RT-qPCR and immunohistochemistry in a retrospective series of HCC surgical samples, and correlated with the presence of mVI. The functional role of hMOF in HCC vascular invasion was investigated in vitro in HCC cell lines using siRNA, transcriptomic analysis and transwell invasion assay, and in vivo using a Zebrafish embryo xenograft model. RESULTS We found that hMOF was significantly upregulated at the protein level in HCC with mVI, compared with HCC without mVI (P < .01). Transcriptomic analysis showed that hMOF downregulation in HCC cell line lead to significant downregulation of key genes and pathways involved in vascular invasion. These results were confirmed by transwell invasion assay, where hMOF downregulation significantly reduced HCC cells invasion. Finally, hMOF downregulation significantly reduced tumour cell intravasation and metastasis in vivo. CONCLUSIONS Altogether, these results underpin a critical role for hMOF in vascular invasion in HCC, via transcription activation of key genes involved in this process. These data confirm the major role of epigenetic alterations in HCC progression, and pave the way for future therapies targeting hMOF in HCC.
Collapse
Affiliation(s)
- Nicolas Poté
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| | - Jérôme Cros
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| | - Samira Laouirem
- INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| | - Jérôme Raffenne
- INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| | - Magda Negrão
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
| | - Miguel Albuquerque
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Pierre Bedossa
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| | - Miguel Godinho Ferreira
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal.,INSERM U1081 UMR7284 CNRS, Institute for Research on Cancer and Aging of Nice (IRCAN), Nice, France
| | - Slimane Ait Si Ali
- Centre Epigénétique et Destin Cellulaire, UMR7216, CNRS, Paris Diderot University, Sorbonne Paris Cité, Paris, France
| | - Rita Fior
- Champalimaud Centre for the Unknown, Champalimaud Foundation, Lisbon, Portugal
| | - Valérie Paradis
- Department of Pathology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,INSERM UMR 1149, Centre de recherche sur l'inflammation, Paris, France
| |
Collapse
|
11
|
Naqvi MAUH, Memon MA, Jamil T, Naqvi SZ, Aimulajiang K, Gadahi JA, Xu L, Song X, Li X, Yan R. Galectin Domain Containing Protein from Haemonchus contortus Modulates the Immune Functions of Goat PBMCs and Regulates CD4+ T-Helper Cells In Vitro. Biomolecules 2020; 10:E116. [PMID: 31936604 PMCID: PMC7022894 DOI: 10.3390/biom10010116] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/05/2020] [Accepted: 01/05/2020] [Indexed: 02/08/2023] Open
Abstract
Galectins are glycan-binding proteins that are widely expressed and distributed in mammalian tissues as well as cells of innate and adaptive immune responses. CD4+ T-helper cells differentiate into effector subsets in response to cytokines. T helper 9 cells are one of the recently described subsets of effector T cells that are relatively new and less studied. In this study, galectin domain containing protein from Haemonchus contortus (Hc-GDC) was cloned, expressed in pET32a, and immunoblotting was performed. Localization of recombinant (r)Hc-GDC on outer and inner surface of H. contortus worm and binding with goat Peripheral Blood Mononuclear cells (PBMCs) were performed using immunofluorescence assay. Moreover, effects of rHc-GDC on proliferation, apoptosis, cell migration, and the nitric oxide production in goat PBMCs were evaluated. Furthermore, modulatory effects of rHc-GDC on production of Th1, Th2, and Th9 cells were evaluated by flowcytometry and on interferon gamma, interleukin (IL)-4 and IL-9 were evaluated by quantitative real-time polymerase chain reaction. The results demonstrated that rHc-GDC was successfully cloned, expressed in expression vector as well as in the gut surface of adult H. contortus worm and successful binding with PBMCs surface were observed. Immunoblotting results revealed that rHc-GDC is an important active protein of H. contortus excretory and secretory products. Moreover, the interaction of rHc-GDC with host cells increased the production of Th2, Th9 cells, IL4, IL-9, PBMC proliferation, nitric oxide, and cell migration. No effects of rHc-GDC were observed on PMBC apoptosis, production of Th1 cells, and secretions of IFN- and IL-10 cytokines. These findings indicate that recombinant GDC protein from H. contortus modulates the immune functions of goat PBMCs and has the potential to enhance protective immunity by inducing T helper-9-derived IL-9 in vitro.
Collapse
Affiliation(s)
- Muhammad Ali-ul-Husnain Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Muhammad Ali Memon
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Tahseen Jamil
- Sindh Agriculture University, Tandojam 70050, Sindh, Pakistan; (T.J.); (J.A.G.)
| | - Sana Zahra Naqvi
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Kalibixiati Aimulajiang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Javaid Ali Gadahi
- Sindh Agriculture University, Tandojam 70050, Sindh, Pakistan; (T.J.); (J.A.G.)
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.-u.-H.N.); (M.A.M.); (S.Z.N.); (K.A.); (L.X.); (X.S.); (X.L.)
| |
Collapse
|
12
|
Bacigalupo ML, Piazza VG, Cicconi NS, Carabias P, Bartke A, Fang Y, Sotelo AI, Rabinovich GA, Troncoso MF, Miquet JG. Growth hormone upregulates the pro-tumorigenic galectin 1 in mouse liver. Endocr Connect 2019; 8:1108-1117. [PMID: 31272083 PMCID: PMC6652241 DOI: 10.1530/ec-19-0292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 07/04/2019] [Indexed: 12/24/2022]
Abstract
Transgenic mice overexpressing growth hormone (GH) spontaneously develop liver tumors, including hepatocellular carcinoma (HCC), within a year. The preneoplastic liver pathology in these mice recapitulates that observed in humans at high risk of developing hepatic cancer. Although increased expression of galectin 1 (GAL1) in liver tissue is associated with HCC aggressiveness, a link between this glycan-binding protein and hormone-related tumor development has not yet been explored. In this study, we investigated GAL1 expression during liver tumor progression in mice continuously exposed to high levels of GH. GAL1 expression was determined by Western blotting, RT-qPCR and immunohistochemistry in the liver of transgenic mice overexpressing GH. Animals of representative ages at different stages of liver pathology were studied. GAL1 expression was upregulated in the liver of GH-transgenic mice. This effect was observed at early ages, when animals displayed no signs of liver disease or minimal histopathological alterations and was also detected in young adults with preneoplastic liver pathology. Remarkably, GAL1 upregulation was sustained during aging and its expression was particularly enhanced in liver tumors. GH also induced hepatic GAL1 expression in mice that were treated with this hormone for a short period. Moreover, GH triggered a rapid increment in GAL1 protein expression in human HCC cells, denoting a direct effect of the hormone on hepatocytes. Therefore, our results indicate that GH upregulates GAL1 expression in mouse liver, which may have critical implications in tumorigenesis. These findings suggest that this lectin could be implicated in hormone-driven liver carcinogenesis.
Collapse
Affiliation(s)
- María L Bacigalupo
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Verónica G Piazza
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Nadia S Cicconi
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Pablo Carabias
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Andrzej Bartke
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Yimin Fang
- Department of Internal Medicine, Geriatrics Research, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Ana I Sotelo
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas, and Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María F Troncoso
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| | - Johanna G Miquet
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Química y Fisicoquímica Biológicas, Buenos Aires, Argentina
| |
Collapse
|
13
|
Potikha T, Pappo O, Mizrahi L, Olam D, Maller SM, Rabinovich GA, Galun E, Goldenberg DS. Lack of galectin-1 exacerbates chronic hepatitis, liver fibrosis, and carcinogenesis in murine hepatocellular carcinoma model. FASEB J 2019; 33:7995-8007. [PMID: 30897344 PMCID: PMC9292271 DOI: 10.1096/fj.201900017r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/18/2019] [Indexed: 04/16/2024]
Abstract
Chronic liver inflammation (CLI) is a risk factor for development of hepatocellular carcinoma (HCC). Galectin-1 (Gal1) is involved in the regulation of inflammation, angiogenesis, and tumorigenesis, exhibiting multiple anti-inflammatory and protumorigenic activities. We aimed to explore its regulatory role in CLI and HCC progression using an established model of CLI-mediated HCC development, Abcb4 [multidrug-resistance 2 (Mdr2)]-knockout (KO) mice, which express high levels of Gal1 in the liver. We generated double-KO (dKO) Gal1-KO/Mdr2-KO mice on C57BL/6 and FVB/N genetic backgrounds and compared HCC development in the generated strains with their parental Mdr2-KO strains. Loss of Gal1 increased liver injury, inflammation, fibrosis, and ductular reaction in dKO mice of both strains starting from an early age. Aged dKO mutants displayed earlier hepatocarcinogenesis and increased tumor size compared with control Mdr2-KO mice. We found that osteopontin, a well-known modulator of HCC development, and oncogenic proteins Ntrk2 (TrkB) and S100A4 were overexpressed in dKO compared with Mdr2-KO livers. Our results demonstrate that in Mdr2-KO mice, a model of CLI-mediated HCC, Gal1-mediated protection from hepatitis, liver fibrosis, and HCC initiation dominates over its known procarcinogenic activities at later stages of HCC development. These findings suggest that anti-Gal1 treatments may not be applicable at all stages of CLI-mediated HCC.-Potikha, T., Pappo, O., Mizrahi, L., Olam, D., Maller, S. M., Rabinovich, G. A., Galun, E., Goldenberg, D. S. Lack of galectin-1 exacerbates chronic hepatitis, liver fibrosis, and carcinogenesis in murine hepatocellular carcinoma model.
Collapse
Affiliation(s)
- Tamara Potikha
- The Goldyne Savad Institute of Gene TherapyHadassah-Hebrew University Medical CenterJerusalemIsrael
| | - Orit Pappo
- Department of PathologyHadassah-Hebrew University Medical CenterJerusalemIsrael
| | - Lina Mizrahi
- The Goldyne Savad Institute of Gene TherapyHadassah-Hebrew University Medical CenterJerusalemIsrael
| | - Devorah Olam
- The Goldyne Savad Institute of Gene TherapyHadassah-Hebrew University Medical CenterJerusalemIsrael
| | - Sebastián M. Maller
- Laboratory of ImmunopathologyInstitute of Biology and Experimental Medicine (IBYME)Argentinean National Research Council (CONICET)Buenos AiresArgentina
| | - Gabriel A. Rabinovich
- Laboratory of ImmunopathologyInstitute of Biology and Experimental Medicine (IBYME)Argentinean National Research Council (CONICET)Buenos AiresArgentina
- Faculty of Exact and Natural SciencesUniversity of Buenos AiresBuenos AiresArgentina
| | - Eithan Galun
- The Goldyne Savad Institute of Gene TherapyHadassah-Hebrew University Medical CenterJerusalemIsrael
| | - Daniel S. Goldenberg
- The Goldyne Savad Institute of Gene TherapyHadassah-Hebrew University Medical CenterJerusalemIsrael
| |
Collapse
|
14
|
Dubé-Delarosbil C, St-Pierre Y. The emerging role of galectins in high-fatality cancers. Cell Mol Life Sci 2018; 75:1215-1226. [PMID: 29119229 PMCID: PMC11105754 DOI: 10.1007/s00018-017-2708-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/16/2017] [Accepted: 11/02/2017] [Indexed: 12/13/2022]
Abstract
Although we witnessed considerable progress in the prevention and treatment of cancer during the past few decades, a number of cancers remain difficult to treat. The main reasons for this are a lack of effective biomarkers necessary for an early detection and inefficient treatments for cancer that are diagnosed at late stages of the disease. Because of their alarmin-like properties and their protumorigenic role during cancer progression, members of the galectin family are uniquely positioned to provide information that could be used for the exploration of possible avenues for the treatment of high fatality cancer (HFC). A rapid overview of studies that examined the expressions and functions of galectins in cancer cells reveals that they play a central role in at least three major features that characterize HFCs: (1) induction of systemic and local immunosuppression, (2) chemoresistance of cancer cells, and (3) increased invasive behavior. Defining the galectinome in HFCs will also lead to a better understanding of tumor heterogeneity while providing critical information that could improve the accuracy of biomarker panels for a more personalized treatment of HFCs. In this review, we discuss the relevance of the galectinome in HFC and its possible contribution to providing potential solutions.
Collapse
Affiliation(s)
| | - Yves St-Pierre
- INRS-Institut Armand-Frappier, 531 Boul. des Prairies, Laval, QC, H7V 1B7, Canada.
| |
Collapse
|
15
|
Immune Evasion in Pancreatic Cancer: From Mechanisms to Therapy. Cancers (Basel) 2018; 10:cancers10010006. [PMID: 29301364 PMCID: PMC5789356 DOI: 10.3390/cancers10010006] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA), the most frequent type of pancreatic cancer, remains one of the most challenging problems for the biomedical and clinical fields, with abysmal survival rates and poor therapy efficiency. Desmoplasia, which is abundant in PDA, can be blamed for much of the mechanisms behind poor drug performance, as it is the main source of the cytokines and chemokines that orchestrate rapid and silent tumor progression to allow tumor cells to be isolated into an extensive fibrotic reaction, which results in inefficient drug delivery. However, since immunotherapy was proclaimed as the breakthrough of the year in 2013, the focus on the stroma of pancreatic cancer has interestingly moved from activated fibroblasts to the immune compartment, trying to understand the immunosuppressive factors that play a part in the strong immune evasion that characterizes PDA. The PDA microenvironment is highly immunosuppressive and is basically composed of T regulatory cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressive cells (MDSCs), which block CD8⁺ T-cell duties in tumor recognition and clearance. Interestingly, preclinical data have highlighted the importance of this immune evasion as the source of resistance to single checkpoint immunotherapies and cancer vaccines and point at pathways that inhibit the immune attack as a key to solve the therapy puzzle. Here, we will discuss the molecular mechanisms involved in PDA immune escape as well as the state of the art of the PDA immunotherapy.
Collapse
|
16
|
Potikha T, Ella E, Cerliani JP, Mizrahi L, Pappo O, Rabinovich GA, Galun E, Goldenberg DS. Galectin-1 is essential for efficient liver regeneration following hepatectomy. Oncotarget 2017; 7:31738-54. [PMID: 27166189 PMCID: PMC5077973 DOI: 10.18632/oncotarget.9194] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/19/2016] [Indexed: 01/23/2023] Open
Abstract
Galectin-1 (Gal1) is a known immune/inflammatory regulator which acts both extracellularly and intracellularly, modulating innate and adaptive immune responses. Here, we explored the role of Gal1 in liver regeneration using 70% partial hepatectomy (PHx) of C57BL/6 wild type and Gal1-knockout (Gal1-KO, Lgals1−/−) mice. Gene or protein expression, in liver samples collected at time intervals from 2 to 168 hours post-operation, was tested by either RT-PCR or by immunoblotting and immunohistochemistry, respectively. We demonstrated that Gal1 transcript and protein expression was induced in the liver tissue of wild type mice upon PHx. Liver regeneration following PHx was significantly delayed in the Gal1-KO compared to the control liver. This delay was accompanied by a decreased Akt phosphorylation, and accumulation of the hepatocyte nuclear p21 protein in the Gal1-KO versus control livers at 24 and 48 hours following PHx. Transcripts of several known regulators of inflammation, cell cycle and cell signaling, including some known PHx-induced genes, were aberrantly expressed (mainly down-regulated) in Gal1-KO compared to control livers at 2, 6 and 24 hours post-PHx. Transient steatosis, which is imperative for liver regeneration following PHx, was significantly delayed and decreased in the Gal1-KO compared to the control liver and was accompanied by a significantly decreased expression in the mutant liver of several genes encoding lipid metabolism regulators. Our results demonstrate that Gal1 protein is essential for efficient liver regeneration following PHx through the regulation of liver inflammation, hepatic cell proliferation, and the control of lipid storage in the regenerating liver.
Collapse
Affiliation(s)
- Tamara Potikha
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ezra Ella
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Juan P Cerliani
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, CONICET, Buenos Aires, Argentina
| | - Lina Mizrahi
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, Institute of Biology and Experimental Medicine, CONICET, Buenos Aires, Argentina.,Faculty of Exact and Natural Sciences, University of Buenos Aires, Buenos Aires, Argentina
| | - Eithan Galun
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniel S Goldenberg
- The Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
17
|
Bacigalupo ML, Carabias P, Troncoso MF. Contribution of galectin-1, a glycan-binding protein, to gastrointestinal tumor progression. World J Gastroenterol 2017; 23:5266-5281. [PMID: 28839427 PMCID: PMC5550776 DOI: 10.3748/wjg.v23.i29.5266] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 05/04/2017] [Accepted: 06/18/2017] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal cancer is a group of tumors that affect multiple sites of the digestive system, including the stomach, liver, colon and pancreas. These cancers are very aggressive and rapidly metastasize, thus identifying effective targets is crucial for treatment. Galectin-1 (Gal-1) belongs to a family of glycan-binding proteins, or lectins, with the ability to cross-link specific glycoconjugates. A variety of biological activities have been attributed to Gal-1 at different steps of tumor progression. Herein, we summarize the current literature regarding the roles of Gal-1 in gastrointestinal malignancies. Accumulating evidence shows that Gal-1 is drastically up-regulated in human gastric cancer, hepatocellular carcinoma, colorectal cancer and pancreatic ductal adenocarcinoma tissues, both in tumor epithelial and tumor-associated stromal cells. Moreover, Gal-1 makes a crucial contribution to the pathogenesis of gastrointestinal malignancies, favoring tumor development, aggressiveness, metastasis, immunosuppression and angiogenesis. We also highlight that alterations in Gal-1-specific glycoepitopes may be relevant for gastrointestinal cancer progression. Despite the findings obtained so far, further functional studies are still required. Elucidating the precise molecular mechanisms modulated by Gal-1 underlying gastrointestinal tumor progression, might lead to the development of novel Gal-1-based diagnostic methods and/or therapies.
Collapse
|
18
|
Quantitative proteomics reveal the anti-tumour mechanism of the carbohydrate recognition domain of Galectin-3 in Hepatocellular carcinoma. Sci Rep 2017; 7:5189. [PMID: 28701735 PMCID: PMC5507876 DOI: 10.1038/s41598-017-05419-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 05/30/2017] [Indexed: 01/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a serious threat to human health. The carbohydrate recognition domain of Galectin-3 (Gal3C) has been reported to be an anti-tumour molecule. In this study, we aim to explore effects of Gal3C in HCC and its possible molecular mechanism with quantitative proteomics approach. We found that rGal3C stimulation could inhibit cell viability, migration and invasion of HepG2. After rGal3C stimulating, 190 proteins were differentially expressed. Eighty up-regulated proteins located mainly in extracellular exosome and involved in cell adhesion and metabolism, and 110 down-regulated proteins located in mitochondria and extracellular exosome, and related to processes of metabolism and oxidation-reduction. Of the differentially expressed proteins, CLU, NDRG1, CD166, S100A11 and Galectin-1 were carcinoma-related proteins affected by rGal3C. Potential receptors of rGal3C were explored by an UV cross-linking capture strategy. We showed that rGal3C could induce dephosphorylating of FAK/SRC. Blocking of the FAK/SRC pathway resulted in down-regulation of NDRG1. Immunofluorescence suggested that rGal3C could disrupt integrin clustering. Our study provides valuable insight into the anti-tumour mechanism of rGal3C in HCC on a proteomics level and is the first to reveal the possible mechanism involving integrin/FAK/SRC pathway and NDRG1. These results provide useful guidance of developing new therapies for HCC.
Collapse
|
19
|
Fernández MM, Ferragut F, Cárdenas Delgado VM, Bracalente C, Bravo AI, Cagnoni AJ, Nuñez M, Morosi LG, Quinta HR, Espelt MV, Troncoso MF, Wolfenstein-Todel C, Mariño KV, Malchiodi EL, Rabinovich GA, Elola MT. Glycosylation-dependent binding of galectin-8 to activated leukocyte cell adhesion molecule (ALCAM/CD166) promotes its surface segregation on breast cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1860:2255-2268. [PMID: 27130882 DOI: 10.1016/j.bbagen.2016.04.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 03/27/2016] [Accepted: 04/23/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND We previously demonstrated that the activated leukocyte cell adhesion molecule (ALCAM/CD166) can interact with galectin-8 (Gal-8) in endothelial cells. ALCAM is a member of the immunoglobulin superfamily that promotes homophilic and heterophilic cell-cell interactions. Gal-8 is a "tandem-repeat"-type galectin, known as a matricellular protein involved in cell adhesion. Here, we analyzed the physical interaction between both molecules in breast cancer cells and the functional relevance of this phenomenon. METHODS We performed binding assays by surface plasmon resonance to study the interaction between Gal-8 and the recombinant glycosylated ALCAM ectodomain or endogenous ALCAM from MDA-MB-231 breast cancer cells. We also analyzed the binding of ALCAM-silenced or control breast cancer cells to immobilized Gal-8 by SPR. In internalization assays, we evaluated the influence of Gal-8 on ALCAM surface localization. RESULTS We showed that recombinant glycosylated ALCAM and endogenous ALCAM from breast carcinoma cells physically interacted with Gal-8 in a glycosylation-dependent fashion displaying a differential behavior compared to non-glycosylated ALCAM. Moreover, ALCAM-silenced breast cancer cells exhibited reduced binding to Gal-8 relative to control cells. Importantly, exogenously added Gal-8 provoked ALCAM segregation, probably trapping this adhesion molecule at the surface of breast cancer cells. CONCLUSIONS Our data indicate that Gal-8 interacts with ALCAM at the surface of breast cancer cells through glycosylation-dependent mechanisms. GENERAL SIGNIFICANCE A novel heterophilic interaction between ALCAM and Gal-8 is demonstrated here, suggesting its physiologic relevance in the biology of breast cancer cells.
Collapse
Affiliation(s)
- Marisa M Fernández
- Institute of Studies in Humoral Immunology, University of Buenos Aires (UBA) and National Council Research (CONICET), Microbiology, Immunology and Biotechnology Department, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Fátima Ferragut
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Víctor M Cárdenas Delgado
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Candelaria Bracalente
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Alicia I Bravo
- Molecular Pathology Department, "Eva Perón" HIGA Hospital, Buenos Aires, Argentina
| | - Alejandro J Cagnoni
- Laboratory of Functional and Molecular Glycomics, Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina
| | - Myriam Nuñez
- Department of Mathematics and Statistics, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Luciano G Morosi
- Laboratory of Functional and Molecular Glycomics, Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina; Laboratory of Immunopathology, IBYME, CONICET, Buenos Aires, Argentina
| | - Héctor R Quinta
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - María V Espelt
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - María F Troncoso
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Carlota Wolfenstein-Todel
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina
| | - Karina V Mariño
- Laboratory of Functional and Molecular Glycomics, Institute of Biology and Experimental Medicine (IBYME), CONICET, Buenos Aires, Argentina
| | - Emilio L Malchiodi
- Institute of Studies in Humoral Immunology, University of Buenos Aires (UBA) and National Council Research (CONICET), Microbiology, Immunology and Biotechnology Department, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Laboratory of Immunopathology, IBYME, CONICET, Buenos Aires, Argentina; Faculty of Exact and Natural Sciences, UBA, Buenos Aires, Argentina
| | - María T Elola
- Institute of Biochemistry and Biophysics (IQUIFIB), UBA-CONICET, Biological Chemistry Department, School of Pharmacy and Biochemistry, UBA, Buenos Aires, Argentina.
| |
Collapse
|
20
|
Manzi M, Bacigalupo ML, Carabias P, Elola MT, Wolfenstein-Todel C, Rabinovich GA, Espelt MV, Troncoso MF. Galectin-1 Controls the Proliferation and Migration of Liver Sinusoidal Endothelial Cells and Their Interaction With Hepatocarcinoma Cells. J Cell Physiol 2016; 231:1522-1533. [PMID: 26551914 DOI: 10.1002/jcp.25244] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 10/22/2015] [Indexed: 01/05/2023]
Abstract
Galectin-1 (Gal1), a β-galactoside-binding protein elevated in hepatocellular carcinoma (HCC), promotes epithelial-mesenchymal transition (EMT) and its expression correlates with HCC growth, invasiveness, and metastasis. During the early stages of HCC, transforming growth factor β1 (TGF-β1 ) acts as a tumor suppressor; however in advanced stages, HCC cells lose their cytostatic response to TGF-β1 and undergo EMT. Here, we investigated the role of Gal1 on liver endothelial cell biology, and the interplay between Gal1 and TGF-β1 in HCC progression. By Western blot and immunofluorescence, we analyzed Gal1 expression, secretion and localization in HepG2 and HuH-7 human HCC cells, and in SK-HEP-1 human liver sinusoidal endothelial cells (SECs). We used loss-of-function and gain-of-function experiments to down- or up-regulate Gal1 expression, respectively, in HepG2 cells. We cultured SK-HEP-1 cells with conditioned media from HCC cells secreting different levels of Gal1, and demonstrated that Gal1 derived from tumor hepatocytes induced its own expression in SECs. Colorimetric and scratch-wound assays revealed that secretion of Gal1 by HCC cells induced SEC proliferation and migration. Moreover, by fluorescence microscopy we demonstrated that Gal1 promoted glycan-dependent heterotypic adhesion of HepG2 cells to SK-HEP-1 SECs. Furthermore, TGF-β1 induced Gal1 expression and secretion by HCC cells, and promoted HepG2 cell adhesion to SK-HEP-1 SECs through a Gal1-dependent mechanism. Finally, Gal1 modulated HepG2 cell proliferation and sensitivity to TGF-β1 -induced growth inhibition. Our results suggest that Gal1 and TGF-β1 might function coordinately within the HCC microenvironment to regulate tumor growth, invasion, metastasis, and angiogenesis.
Collapse
Affiliation(s)
- Malena Manzi
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - María L Bacigalupo
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Pablo Carabias
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - María T Elola
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Carlota Wolfenstein-Todel
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - Gabriel A Rabinovich
- Institute of Biology and Experimental Medicine (CONICET) and School of Exact and Natural Sciences, University of Buenos Aires (UBA), Buenos Aires, Argentina
| | - María V Espelt
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| | - María F Troncoso
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires (UBA), Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini", University of Buenos Aires (UBA)-National Research Council of Argentina (CONICET), Buenos Aires, Argentina
| |
Collapse
|
21
|
Su YC, Davuluri GVN, Chen CH, Shiau DC, Chen CC, Chen CL, Lin YS, Chang CP. Galectin-1-Induced Autophagy Facilitates Cisplatin Resistance of Hepatocellular Carcinoma. PLoS One 2016; 11:e0148408. [PMID: 26859293 PMCID: PMC4747500 DOI: 10.1371/journal.pone.0148408] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/17/2016] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers in Taiwan. Although chemotherapy is the primary treatment for HCC patients, drug resistance often leads to clinical failure. Galectin-1 is a beta-galactoside binding lectin which is up-regulated in HCC patients and promotes tumor growth by mediating cancer cell adhesion, migration and proliferation, but its role in chemoresistance of HCC is poorly understood. In this study we found that galectin-1 is able to lead to chemoresistance against cisplatin treatment, and subsequent inhibition has reversed the effect of cell death in HCC cells. Moreover, galectin-1 was found to induce autophagic flux in HCC cells. Inhibition of autophagy by inhibitors or knockdown of Atg5 cancels galectin-1-induced cisplatin resistance in HCC cells. Increase of mitophagy triggered by galectin-1 was found to reduce the mitochondrial potential loss and apoptosis induced by cisplatin treatment. Finally, using an in situ hepatoma mouse model, we clearly demonstrated that inhibition of galectin-1 by thiodigalactoside could significantly augment the anti-HCC effect of cisplatin. Taken together, our findings offer a new insight into the chemoresistance galectin-1 causes against cisplatin treatment, and points to a potential approach to improve the efficacy of cisplatin in the treatment of HCC patients.
Collapse
Affiliation(s)
- Yu-Chi Su
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | | | - Cheng-Hao Chen
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Dong-Che Shiau
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chien-Chin Chen
- Department of Pathology, Chia-Yi Christian Hospital, Chiayi 600, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan 701, Taiwan
| | - Chia-Ling Chen
- Translational Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Yee-Shin Lin
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan
| | - Chih-Peng Chang
- Department of Microbiology & Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- The Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan
- * E-mail:
| |
Collapse
|
22
|
Li J, Sun RR, Yu ZJ, Liang H, Shen S, Kan Q. Galectin-1 Modulates the Survival and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) Sensitivity in Human Hepatocellular Carcinoma Cells. Cancer Biother Radiopharm 2015; 30:336-41. [PMID: 26348206 DOI: 10.1089/cbr.2015.1857] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Juan Li
- Department of Infectious Disease, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Ran-ran Sun
- Department of Infectious Disease, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Zu-jiang Yu
- Department of Infectious Disease, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Hongxia Liang
- Department of Infectious Disease, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Shen Shen
- Department of Infectious Disease, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| | - Quancheng Kan
- Department of Pharmacy, The First Affiliated Hospital of ZhengZhou University, ZhengZhou, China
| |
Collapse
|
23
|
Restuccia A, Tian YF, Collier JH, Hudalla GA. Self-assembled glycopeptide nanofibers as modulators of galectin-1 bioactivity. Cell Mol Bioeng 2015; 8:471-487. [PMID: 26495044 DOI: 10.1007/s12195-015-0399-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Galectins are carbohydrate-binding proteins that act as extracellular signaling molecules in various normal and pathological processes. Galectin bioactivity is mediated by specific non-covalent interactions with cell-surface and extracellular matrix (ECM) glycoproteins, which can enhance or inhibit signaling events that influence various cellular behaviors, including adhesion, proliferation, differentiation, and apoptosis. Here, we developed a materials approach to modulate galectin bioactivity by mimicking natural galectin-glycoprotein interactions. Specifically, we created a variant of a peptide that self-assembles into β-sheet nanofibers under aqueous conditions, QQKFQFQFEQQ (Q11), which has an asparagine residue modified with the monosaccharide N-acetylglucosamine (GlcNAc) at its N-terminus (GlcNAc-Q11). GlcNAc-Q11 self-assembled into β-sheet nanofibers under similar conditions as Q11. Nanofibrillar GlcNAc moieties were efficiently converted to the galectin-binding disaccharide N-acetyllactosamine (LacNAc) via the enzyme β-1,4-galactosyltransferase and the sugar donor UDP-galactose, while retaining β-sheet structure and nanofiber morphology. LacNAc-Q11 nanofibers bound galectin-1 and -3 in a LacNAc concentration-dependent manner, although nanofibers bound galectin-1 with higher affinity than galectin-3. In contrast, galectin-1 bound weakly to GlcNAc-Q11 nanofibers, while no galectin-3 binding to these nanofibers was observed. Galectin-1 binding to LacNAc-Q11 nanofibers was specific because it could be inhibited by excess soluble β-lactose, a galectin-binding carbohydrate. LacNAc-Q11 nanofibers inhibited galectin-1-mediated apoptosis of Jurkat T cells in a LacNAc concentration-dependent manner, but were unable to inhibit galectin-3 activity, consistent with galectin-binding affinity of the nanofibers. We envision that glycopeptide nanofibers capable of modulating galectin-1 bioactivity will be broadly useful as biomaterials for various medical applications, including cancer therapeutics, immunotherapy, tissue regeneration, and viral prophylaxis.
Collapse
Affiliation(s)
| | - Ye F Tian
- Department of Surgery, University of Chicago. ; Department of Biomedical Engineering, Illinois Institute of Technology
| | | | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering. ; Department of Surgery, University of Chicago
| |
Collapse
|
24
|
Bacigalupo ML, Manzi M, Espelt MV, Gentilini LD, Compagno D, Laderach DJ, Wolfenstein-Todel C, Rabinovich GA, Troncoso MF. Galectin-1 triggers epithelial-mesenchymal transition in human hepatocellular carcinoma cells. J Cell Physiol 2015; 230:1298-1309. [PMID: 25469885 DOI: 10.1002/jcp.24865] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 11/06/2014] [Indexed: 12/23/2022]
Abstract
Galectin-1 (Gal1), a β-galactoside-binding protein abundantly expressed in tumor microenvironments, is associated with the development of metastasis in hepatocellular carcinomas (HCC). However, the precise roles of Gal1 in HCC cell invasiveness and dissemination are uncertain. Here, we investigated whether Gal1 mediate epithelial-mesenchymal transition (EMT) in HCC cells, a key process during cancer progression. We used the well-differentiated and low invasive HepG2 cells and performed 'gain-of-function' and 'loss-function' experiments by transfecting cells with Gal1 cDNA constructs or by siRNA strategies, respectively. Epithelial and mesenchymal markers expression, changes in apico-basal polarity, independent-anchorage growth, and activation of specific signaling pathways were studied using Western blot, fluorescence microscopy, soft-agar assays, and FOP/TOP flash reporter system. Gal1 up-regulation in HepG2 cells induced down-regulation of the adherens junction protein E-cadherin and increased expression of the transcription factor Snail, one of the main inducers of EMT in HCC. Enhanced Gal1 expression facilitated the transition from epithelial cell morphology towards a fibroblastoid phenotype and favored up-regulation of the mesenchymal marker vimentin in HCC cells. Cells overexpressing Gal1 showed enhanced anchorage-independent growth and loss of apico-basal polarity. Remarkably, Gal1 promoted Akt activation, β-catenin nuclear translocation, TCF4/LEF1 transcriptional activity and increased cyclin D1 and c-Myc expression, suggesting activation of the Wnt pathway. Furthermore, Gal1 overexpression induced E-cadherin downregulation through a PI3K/Akt-dependent mechanism. Our results provide the first evidence of a role of Gal1 as an inducer of EMT in HCC cells, with critical implications in HCC metastasis.
Collapse
Affiliation(s)
- María L Bacigalupo
- Institute of Biological Chemistry and Biophysics "Prof. Alejandro C. Paladini" (UBA-CONICET), Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
During the past decade, a better understanding of the cellular and molecular mechanisms underlying tumor immunity has provided the appropriate framework for the development of therapeutic strategies for cancer immunotherapy. Under this complex scenario, galectins have emerged as promising molecular targets for cancer therapy responsible of creating immunosuppressive microenvironments at sites of tumor growth and metastasis. Galectins, expressed in tumor, stromal, and endothelial cells, contribute to thwart the development of immune responses by favoring the expansion of T regulatory cells and contributing to their immunosuppressive activity, driving the differentiation of tolerogenic dendritic cells, limiting T cell viability, and maintaining T cell anergy. The emerging data promise a future scenario in which the selective blockade of individual members of the galectin family, either alone or in combination with other therapeutic regimens, will contribute to halt tumor progression by counteracting tumor-immune escape. Here we describe a selection of methods used to investigate the role of galectin-1 in tumor-immune escape.
Collapse
|
26
|
Yeh CC, Hsu CH, Shao YY, Ho WC, Tsai MH, Feng WC, Chow LP. Integrated Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) and Isobaric Tags for Relative and Absolute Quantitation (iTRAQ) Quantitative Proteomic Analysis Identifies Galectin-1 as a Potential Biomarker for Predicting Sorafenib Resistance in Liver Cancer. Mol Cell Proteomics 2015; 14:1527-45. [PMID: 25850433 DOI: 10.1074/mcp.m114.046417] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Indexed: 01/06/2023] Open
Abstract
Sorafenib has become the standard therapy for patients with advanced hepatocellular carcinoma (HCC). Unfortunately, most patients eventually develop acquired resistance. Therefore, it is important to identify potential biomarkers that could predict the efficacy of sorafenib. To identify target proteins associated with the development of sorafenib resistance, we applied stable isotope labelling with amino acids in cell culture (SILAC)-based quantitative proteomic approach to analyze differences in protein expression levels between parental HuH-7 and sorafenib-acquired resistance HuH-7 (HuH-7(R)) cells in vitro, combined with an isobaric tags for relative and absolute quantitation (iTRAQ) quantitative analysis of HuH-7 and HuH-7(R) tumors in vivo. In total, 2,450 quantified proteins were identified in common in SILAC and iTRAQ experiments, with 81 showing increased expression (>2.0-fold) with sorafenib resistance and 75 showing decreased expression (<0.5-fold). In silico analyses of these differentially expressed proteins predicted that 10 proteins were related to cancer with involvements in cell adhesion, migration, and invasion. Knockdown of one of these candidate proteins, galectin-1, decreased cell proliferation and metastasis in HuH-7(R) cells and restored sensitivity to sorafenib. We verified galectin-1 as a predictive marker of sorafenib resistance and a downstream target of the AKT/mTOR/HIF-1α signaling pathway. In addition, increased galectin-1 expression in HCC patients' serum was associated with poor tumor control and low response rate. We also found that a high serum galectin-1 level was an independent factor associated with poor progression-free survival and overall survival. In conclusion, these results suggest that galectin-1 is a possible biomarker for predicting the response of HCC patients to treatment with sorafenib. As such, it may assist in the stratification of HCC and help direct personalized therapy.
Collapse
Affiliation(s)
- Chao-Chi Yeh
- From the ‡Graduate Institute of Biochemistry and Molecular Biology
| | - Chih-Hung Hsu
- §Graduate Institute of Oncology, College of Medicine, ‖Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Yun Shao
- §Graduate Institute of Oncology, College of Medicine, ‖Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Ching Ho
- From the ‡Graduate Institute of Biochemistry and Molecular Biology
| | - Mong-Hsun Tsai
- ¶Institute of Biotechnology, National Taiwan University and
| | - Wen-Chi Feng
- From the ‡Graduate Institute of Biochemistry and Molecular Biology
| | - Lu-Ping Chow
- From the ‡Graduate Institute of Biochemistry and Molecular Biology,
| |
Collapse
|
27
|
Cuestas ML, Oubiña JR, Mathet VL. Hepatocellular carcinoma and multidrug resistance: Past, present and new challenges for therapy improvement. World J Pharmacol 2015; 4:96-116. [DOI: 10.5497/wjp.v4.i1.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 10/02/2014] [Accepted: 12/01/2014] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most frequent form of liver cancer and the third most common cause of cancer-related death in the world. The main risk factor worldwide for this type of malignancy is chronic hepatitis caused by hepatitis B virus and hepatitis C virus infections. Advances in early detection and treatment have improved life expectancy of patients with HCC. However, this disorder remains as a disease with poor prognosis. In fact, epidemiological studies have revealed that there is an 8-mo median survival rate in patients, approximately 20% of whom survive one year while only 5% remain alive after three years. Additionally, HCC is particularly difficult to treat because of its high recurrence rate, and its resistance to conventional chemotherapy is due, among other mechanisms, to several members of the ATP-Binding Cassette protein family involved in drug transport being overexpressed. Fortunately, there is evidence that these patients may benefit from alternative molecular-targeted therapies. This manuscript intends to provide further insight into the etiology and molecular mechanisms related to HCC development and the latest therapeutic approaches to treat this malignancy. The development of effective delivery systems of antitumor drugs able to target the liver parenchyma is also assessed. Finally, the prospects in the development of more efficient drug therapies to overcome multidrug resistance are also examined.
Collapse
|
28
|
Wang K, Mao Z, Liu L, Zhang R, Liang Q, Xiong Y, Yuan W, Wei L. Rab17 inhibits the tumourigenic properties of hepatocellular carcinomas via the Erk pathway. Tumour Biol 2015; 36:5815-24. [PMID: 25707355 DOI: 10.1007/s13277-015-3251-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/10/2015] [Indexed: 12/12/2022] Open
Abstract
The small GTPase Rab17 is a member of the Rab family and plays a critical role in the regulation of membrane trafficking polarized eukaryotic cells. However, the role of Rab17 in hepatocellular carcinoma (HCC) is not clear. In the present study, we investigated the role of Rab17 in HCC tumourgenesis. The expressions of Rab17 in non-tumour hepatic tissues and HCCs were detected via immunohistochemistry. Rab17 was found in 31 of 48 (64.6 %) and in 23 of 62 (37.1 %) of non-tumour hepatic tissue samples and HCCs (P = 0.0068), respectively, and there were significant correlations between Rab17 reductions and unfavourable variables including tumour size (P = 0.0171), differentiation level (P = 0.0126), and lymph nodal (P = 0.0044) and distant metastases (P = 0.0047). To elucidate the role of Rab17 in HCC, we generated two Rab17-overexpressing HCC cell lines. Rab17 overexpression significantly inhibited the tumourigenic properties of HCC cells in vitro and in vivo as demonstrated by reduced cell proliferation and migration, elevated G1 arrest, and decreased tumour xenograft growth. However, the attenuated tumourigenic properties of the HCC cells that were induced by Rab17 overexpression were significantly rescued by the activator of the Erk pathway EGF, which indicates that the Erk pathway plays a critical role in the Rab17 up-regulation-induced reduced tumourigenic properties of HCC cells. Rab17 might act as a tumour suppressor gene in HCCs, and the anti-tumour effects of Rab17 might be partially mediated by the Erk pathway.
Collapse
Affiliation(s)
- Kejia Wang
- Department of Pathology, No. 401 Hospital of PLA, Qingdao, Shandong, 266071, China
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Jia H, Aw W, Saito K, Hanate M, Hasebe Y, Kato H. Eggshell membrane ameliorates hepatic fibrogenesis in human C3A cells and rats through changes in PPARγ-Endothelin 1 signaling. Sci Rep 2014; 4:7473. [PMID: 25503635 PMCID: PMC5378949 DOI: 10.1038/srep07473] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 11/20/2014] [Indexed: 12/22/2022] Open
Abstract
Our previous nutrigenomic findings indicate that eggshell membrane (ESM) may prevent liver fibrosis. Here we investigated the effects and mechanisms underlying ESM intervention against liver injury by using DNA microarray analysis and comparative proteomics. In vitro hydrolyzed ESM attenuated the TGFβ1-induced procollagen production of human hepatocyte C3A cells and inhibited the expression of Endothelin 1 (EDN1) and its two receptors, and extracellular matrix components. In vivo male Wistar rats were allocated into a normal control group, a CCl4 group (hypodermic injection of 50% CCl4 2×/wk) and an ESM group (20 g ESM/kg diet with CCl4 injection) for 7 wks. Dietary ESM ameliorated the elevated activity of ALT/AST, oxidative stress and collagen accumulation in liver, accompanied by the down-regulated expression of Edn1 signaling and notable profibrogenic genes and growth factors as well as peroxisome proliferator-activated receptor gamma (PPARγ). Concomitantly, the decreased expressions of Galectin-1 and Desmin protein in the ESM group indicated the deactivation of hepatic stellate cells (HSCs). Through a multifaceted integrated omics approach, we have demonstrated that ESM can exert an antifibrotic effect by suppressing oxidative stress and promoting collagen degradation by inhibiting HSCs' transformation, potentially via a novel modulation of the PPARγ-Endothelin 1 interaction signaling pathway.
Collapse
Affiliation(s)
- Huijuan Jia
- Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan
| | - Wanping Aw
- Institute of Advanced Biosciences, Keio University, Yamagata, Japan
| | - Kenji Saito
- Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan
| | - Manaka Hanate
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Hisanori Kato
- 1] Corporate Sponsored Research Program "Food for Life, " Organization for Interdisciplinary Research Projects, The University of Tokyo, Tokyo, Japan [2] Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
30
|
MIAO JINHAO, WANG SHUQIANG, ZHANG MINGHUI, YU FENGBIN, ZHANG LEI, YU ZHONGXIANG, KUANG YONG. Knockdown of galectin-1 suppresses the growth and invasion of osteosarcoma cells through inhibition of the MAPK/ERK pathway. Oncol Rep 2014; 32:1497-504. [PMID: 25069486 DOI: 10.3892/or.2014.3358] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/08/2014] [Indexed: 11/05/2022] Open
|
31
|
Wang W, Wang S, Zhang H, Yuan C, Yan R, Song X, Xu L, Li X. Galectin Hco-gal-m from Haemonchus contortus modulates goat monocytes and T cell function in different patterns. Parasit Vectors 2014; 7:342. [PMID: 25056558 PMCID: PMC4117971 DOI: 10.1186/1756-3305-7-342] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/06/2014] [Indexed: 12/24/2022] Open
Abstract
Background Monocytes and T cells are two major subpopulations of peripheral blood mononuclear cells (PBMC) and play an essential role in the innate and adaptive immune systems. Different members of the galectin family show multiple and distinct regulatory effects on different cell types. Previous studies have demonstrated that the galectin from Haemonchus contortus (Hco-gal-m) performed immunomodulatory effects on goat PBMC, however, which subpopulation of PBMC is the primary target of Hco-gal-m and whether the immune modulations share the same mechanism remain unclear. Methods In this study, the developmental expression of Hco-gal-m was analyzed by RT-PCR and Western blot analysis. The distribution of Hco-gal-m in adult worm was detected by an immunohistochemical test. The binding activity of the recombinant Hco-gal-m (rHco-gal-m) on goat monocytes and T cells were assessed by flow cytometry. The immunomodulatory effects of Hco-gal-m on cytokine secretion, cell activation and apoptosis were observed by co-incubation of rHco-gal-m with goat monocytes and T cells. Results Hco-gal-m was expressed in L4 as well as adult worms and predominantly localized at the internal surface of the worm guts. rHco-gal-m could bind to both monocytes and T cells. The engagement of rHco-gal-m decreased the production of IL-6, IL-10 and TNF-α in T cells, however, it significantly increased the secretion of IL-10 in monocytes. After rHco-gal-m exposure, the expression of MHC-II on monocytes and that of CD25 on T cells were restricted. Consequently, T cell proliferations were potently inhibited by rHco-gal-m. In addition, rHco-gal-m induced apoptosis in T cells, but not significantly in monocytes. Conclusions Our results indicated that rHco-gal-m modulated goat monocytes and T cell function in different patterns. Electronic supplementary material The online version of this article (doi:10.1186/1756-3305-7-342) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
32
|
Shu Z, Li J, Mu N, Gao Y, Huang T, Zhang Y, Wang Z, Li M, Hao Q, Li W, He L, Zhang C, Zhang W, Xue X, Zhang Y. Expression, purification and characterization of galectin-1 in Escherichia coli. Protein Expr Purif 2014; 99:58-63. [PMID: 24718258 DOI: 10.1016/j.pep.2014.03.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 12/23/2022]
Abstract
As a member of beta-galactoside-binding proteins family, Galectin-1 (Gal-1) contains a single carbohydrate recognition domain, by means of which it can bind glycans both as a monomer and as a homodimer. Gal-1 is implicated in modulating cell-cell and cell-matrix interactions and may act as an autocrine negative growth factor that regulates cell proliferation. Besides, it can also suppress TH1 and TH17 cells by regulating dendritic cell differentiation or suppress inflammation via IL-10 and IL-27. In the present study, Gal-1 monomer and concatemer (Gal-1②), which can resemble Gal-1 homodimer, were expressed in Escherichia coli and their bioactivities were analyzed. The results of this indicate that both Gal-1 and Gal-1② were expressed in E. coli in soluble forms with a purity of over 95% after purifying with ion-exchange chromatography. Clearly, both Gal-1 and Gal-1② can effectively promote erythrocyte agglutination in hemagglutinating activity assays and inhibit Jurkat cell proliferation in MTT assays. All these data demonstrate that bacterially-expressed Gal-1 and Gal-1② have activities similar to those of wild type human Gal-1 whereas the bioactivity of concatemer Gal-1② was stronger than those of the bacterially-expressed and wild type human Gal.
Collapse
Affiliation(s)
- Zhen Shu
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Li
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Nan Mu
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yuan Gao
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Tonglie Huang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ying Zhang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Zenglu Wang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Meng Li
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Qiang Hao
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Weina Li
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Liqing He
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
| | - Cun Zhang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Wei Zhang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xiaochang Xue
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Yingqi Zhang
- State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
33
|
Suzuki O, Abe M. Galectin-1-mediated cell adhesion, invasion and cell death in human anaplastic large cell lymphoma: regulatory roles of cell surface glycans. Int J Oncol 2014; 44:1433-42. [PMID: 24589677 PMCID: PMC4027875 DOI: 10.3892/ijo.2014.2319] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/28/2014] [Indexed: 01/01/2023] Open
Abstract
Galectin-1 is known to be one of the extracellular matrix proteins. To elucidate the biological roles of galectin-1 in cell adhesion and invasion of human anaplastic large cell lymphoma, we performed cell adhesion and invasion assays using the anaplastic large cell lymphoma cell line H-ALCL, which was previously established in our laboratory. From the cell surface lectin array, treatment with neuraminidase from Arthrobacter ureafaciens which cleaves all linkage types of cell surface sialic acid enhanced Arachis hypogaea (PNA), Helix pomatia (HPA) and Phaseolus vulgaris-L (L-PHA) lectin binding reactivity to cell surface of lymphoma cells suggesting that neuraminidase removes cell surface sialic acid. In cell adhesion and invasion assays treatment with neuraminidase markedly enhanced cell adhesion to galectin-1 and decreased cell invasive capacity through galectin-1. α2,6-linked sialic acid may be involved in masking the effect of the interaction between galectin-1 and cell surface glycans. H-ALCL cells expressed the β-galactoside-α2,6-sialyltransferase ST6Gal1. On resialylation assay by recombinant ST6Gal1 with CMP-Neu5Ac, α2,6-resialylation of L-PHA reactive oligosaccharide by ST6Gal1 resulted in inhibition of H-ALCL cell adhesion to galectin-1 compared to the desialylated H-ALCL cells. On knockdown experiments, knockdown of ST6Gal1 dramatically enhanced cell adhesion to galectin-1. N-glycosylation inhibitor swainsonine treatment resulted in enhancement of cell adhesion to galectin-1. In glycomic analysis using the lectin blocking assay treatment with PNA, Artocarpus integrifolia (Jacalin), Glycine max (SBA), Helix pomatia (HPA), Vicia villosa (VVA), Ulex europaeus (UEA-1), Triticum vulgaris (WGA), Canavalia ensiformis (ConA), Phaseolus vulgaris-L (L-PHA), Phaseolus vulgaris-E4 (E-PHA), Datura stramonium (DSA) lectins resulted in modulation of lymphoma cell to galectin-1 suggesting that several types of glycans may regulate cell adhesion to galectin-1 by steric hindrance. The adhesive capacity of H-ALCL cells is regulated by phosphatidylinositol 3 phosphate kinase (PI3K) and actin cytoskeleton, and the invasive capacity of H-ALCL cells is regulated by PI3K, mitogen-activated protein kinase (MAPK), Rho and actin cytoskeleton. Furthermore, galectin-1-induced cell death in H-ALCL cells was accompanied by inhibition of CD45 protein tyrosine phosphatase (PTP) activity. In conclusion, cell adhesion and invasion to galectin-1 appeared to be regulated by cell surface sialylation and N-glycosylation, and galectin-1 regulates cell death through inhibition of CD45 PTP activity of H-ALCL.
Collapse
Affiliation(s)
- Osamu Suzuki
- Department of Diagnostic Pathology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Masafumi Abe
- Department of Diagnostic Pathology, School of Medicine, Fukushima Medical University, Fukushima 960-1295, Japan
| |
Collapse
|
34
|
Zhang P, Shi B, Gao H, Jiang H, Kong J, Yan J, Pan X, Li K, Zhang P, Yao M, Yang S, Gu J, Wang H, Li Z. An EpCAM/CD3 bispecific antibody efficiently eliminates hepatocellular carcinoma cells with limited galectin-1 expression. Cancer Immunol Immunother 2014; 63:121-32. [PMID: 24177984 PMCID: PMC11029305 DOI: 10.1007/s00262-013-1497-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/24/2013] [Indexed: 12/17/2022]
Abstract
There have been several studies suggesting that cancer stem cells (CSCs) contribute to the high rates of recurrence and resistance to therapies observed in hepatocellular carcinoma (HCC). Epithelial cell adhesion molecule (EpCAM) has been demonstrated to be a biomarker of CSCs and a potential therapeutic target in HCC. Here, we prepared two anti-EpCAM monoclonal antibodies (1H8 and 2F2) and an anti-EpCAM bispecific T cell engager (BiTE) 1H8/CD3, which was derived from 1H8, and used them to treat HCC in vitro and in vivo. The results demonstrated that all of the developed anti-EpCAM antibodies specifically bound to EpCAM. Neither anti-EpCAM monoclonal antibody had obvious anti-HCC activities in vitro or in vivo. However, anti-EpCAM BiTE 1H8/CD3 induced strong peripheral blood mononuclear cell-dependent cellular cytotoxicity in Huh-7 and Hep3B cells but not EpCAM-negative SK-Hep-1 cells. Notably, 1H8/CD3 completely inhibited the growth of Huh-7 and Hep3B xenografts in vivo. Treatment of the Huh-7 HCC xenografts with 1H8/CD3 significantly suppressed tumor proliferation and reduced the expression of most CSC biomarkers. Intriguingly, galectin-1 (Gal-1) overexpression inhibited 1H8/CD3-induced lymphocytotoxicity in HCCs while knockdown of Gal-1 increased the lymphocytotoxicity. Collectively, these results indicate that anti-EpCAM BiTE 1H8/CD3 is a promising therapeutic agent for HCC treatment. Gal-1 may contribute to the resistance of HCC cells to 1H8/CD3-induced lysis.
Collapse
Affiliation(s)
- Pengfei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Huiping Gao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Juan Kong
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Jin Yan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Xiaorong Pan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Kesang Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Pengwei Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Ming Yao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Shengli Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Jianren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| | - Hongyang Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Institute, Second Military Medical University, Shanghai, 200438 China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln2200, XieTu Rd, Shanghai, 200032 China
| |
Collapse
|
35
|
Bacigalupo ML, Manzi M, Rabinovich GA, Troncoso MF. Hierarchical and selective roles of galectins in hepatocarcinogenesis, liver fibrosis and inflammation of hepatocellular carcinoma. World J Gastroenterol 2013; 19:8831-8849. [PMID: 24379606 PMCID: PMC3870534 DOI: 10.3748/wjg.v19.i47.8831] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 11/02/2013] [Accepted: 11/18/2013] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) represents a global health problem. Infections with hepatitis B or C virus, non-alcoholic steatohepatitis disease, alcohol abuse, or dietary exposure to aflatoxin are the major risk factors to the development of this tumor. Regardless of the carcinogenic insult, HCC usually develops in a context of cirrhosis due to chronic inflammation and advanced fibrosis. Galectins are a family of evolutionarily-conserved proteins defined by at least one carbohydrate recognition domain with affinity for β-galactosides and conserved sequence motifs. Here, we summarize the current literature implicating galectins in the pathogenesis of HCC. Expression of "proto-type" galectin-1, "chimera-type" galectin-3 and "tandem repeat-type" galectin-4 is up-regulated in HCC cells compared to their normal counterparts. On the other hand, the "tandem-repeat-type" lectins galectin-8 and galectin-9 are down-regulated in tumor hepatocytes. The abnormal expression of these galectins correlates with tumor growth, HCC cell migration and invasion, tumor aggressiveness, metastasis, postoperative recurrence and poor prognosis. Moreover, these galectins have important roles in other pathological conditions of the liver, where chronic inflammation and/or fibrosis take place. Galectin-based therapies have been proposed to attenuate liver pathologies. Further functional studies are required to delineate the precise molecular mechanisms through which galectins contribute to HCC.
Collapse
|
36
|
Abstract
Hepatocytes, like other epithelia, are situated at the interface between the organism's exterior and the underlying internal milieu and organize the vectorial exchange of macromolecules between these two spaces. To mediate this function, epithelial cells, including hepatocytes, are polarized with distinct luminal domains that are separated by tight junctions from lateral domains engaged in cell-cell adhesion and from basal domains that interact with the underlying extracellular matrix. Despite these universal principles, hepatocytes distinguish themselves from other nonstriated epithelia by their multipolar organization. Each hepatocyte participates in multiple, narrow lumina, the bile canaliculi, and has multiple basal surfaces that face the endothelial lining. Hepatocytes also differ in the mechanism of luminal protein trafficking from other epithelia studied. They lack polarized protein secretion to the luminal domain and target single-spanning and glycosylphosphatidylinositol-anchored bile canalicular membrane proteins via transcytosis from the basolateral domain. We compare this unique hepatic polarity phenotype with that of the more common columnar epithelial organization and review our current knowledge of the signaling mechanisms and the organization of polarized protein trafficking that govern the establishment and maintenance of hepatic polarity. The serine/threonine kinase LKB1, which is activated by the bile acid taurocholate and, in turn, activates adenosine monophosphate kinase-related kinases including AMPK1/2 and Par1 paralogues has emerged as a key determinant of hepatic polarity. We propose that the absence of a hepatocyte basal lamina and differences in cell-cell adhesion signaling that determine the positioning of tight junctions are two crucial determinants for the distinct hepatic and columnar polarity phenotypes.
Collapse
Affiliation(s)
- Aleksandr Treyer
- Albert Einstein College of Medicine, Department of Developmental and Molecular Biology, Bronx, New York, USA
| | | |
Collapse
|
37
|
Schmandke A, Schmandke A, Pietro MA, Schwab ME. An open source based high content screening method for cell biology laboratories investigating cell spreading and adhesion. PLoS One 2013; 8:e78212. [PMID: 24205161 PMCID: PMC3804740 DOI: 10.1371/journal.pone.0078212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 09/18/2013] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Adhesion dependent mechanisms are increasingly recognized to be important for a wide range of biological processes, diseases and therapeutics. This has led to a rising demand of pharmaceutical modulators. However, most currently available adhesion assays are time consuming and/or lack sensitivity and reproducibility or depend on specialized and expensive equipment often only available at screening facilities. Thus, rapid and economical high-content screening approaches are urgently needed. RESULTS We established a fully open source high-content screening method for identifying modulators of adhesion. We successfully used this method to detect small molecules that are able to influence cell adhesion and cell spreading of Swiss-3T3 fibroblasts in general and/or specifically counteract Nogo-A-Δ20-induced inhibition of adhesion and cell spreading. The tricyclic anti-depressant clomipramine hydrochloride was shown to not only inhibit Nogo-A-Δ20-induced cell spreading inhibition in 3T3 fibroblasts but also to promote growth and counteract neurite outgrowth inhibition in highly purified primary neurons isolated from rat cerebellum. CONCLUSIONS We have developed and validated a high content screening approach that can be used in any ordinarily equipped cell biology laboratory employing exclusively freely available open-source software in order to find novel modulators of adhesion and cell spreading. The versatility and adjustability of the whole screening method will enable not only centers specialized in high-throughput screens but most importantly also labs not routinely employing screens in their daily work routine to investigate the effects of a wide range of different compounds or siRNAs on adhesion and adhesion-modulating molecules.
Collapse
Affiliation(s)
- Andre Schmandke
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Antonio Schmandke
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Maurianne A. Pietro
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Martin E. Schwab
- Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Potikha T, Stoyanov E, Pappo O, Frolov A, Mizrahi L, Olam D, Shnitzer-Perlman T, Weiss I, Barashi N, Peled A, Sass G, Tiegs G, Poirier F, Rabinovich GA, Galun E, Goldenberg D. Interstrain differences in chronic hepatitis and tumor development in a murine model of inflammation-mediated hepatocarcinogenesis. Hepatology 2013; 58:192-204. [PMID: 23423643 DOI: 10.1002/hep.26335] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 02/10/2013] [Indexed: 02/06/2023]
Abstract
UNLABELLED Chronic inflammation is strongly associated with an increased risk for hepatocellular carcinoma (HCC) development. The multidrug resistance 2 (Mdr2)-knockout (KO) mouse (adenosine triphosphate-binding cassette b4(-/-) ), a model of inflammation-mediated HCC, develops chronic cholestatic hepatitis at an early age and HCC at an adult age. To delineate factors contributing to hepatocarcinogenesis, we compared the severity of early chronic hepatitis and late HCC development in two Mdr2-KO strains: Friend virus B-type/N (FVB) and C57 black 6 (B6). We demonstrated that hepatocarcinogenesis was significantly less efficient in the Mdr2-KO/B6 mice versus the Mdr2-KO/FVB mice; this difference was more prominent in males. Chronic hepatitis in the Mdr2-KO/B6 males was more severe at 1 month of age but was less severe at 3 months of age in comparison with age-matched Mdr2-KO/FVB males. A comparative genome-scale gene expression analysis of male livers of both strains at 3 months of age revealed both common and strain-specific aberrantly expressed genes, including genes associated with the regulation of inflammation, the response to oxidative stress, and lipid metabolism. One of these regulators, galectin-1 (Gal-1), possesses both anti-inflammatory and protumorigenic activities. To study its regulatory role in the liver, we transferred the Gal-1-KO mutation (lectin galactoside-binding soluble 1(-/-) ) from the B6 strain to the FVB strain, and we demonstrated that endogenous Gal-1 protected the liver against concanavalin A-induced hepatitis with the B6 genetic background but not the FVB genetic background. CONCLUSION Decreased chronic hepatitis in Mdr2-KO/B6 mice at the age of 3 months correlated with a significant retardation of liver tumor development in this strain versus the Mdr2-KO/FVB strain. We found candidate factors that may determine strain-specific differences in the course of chronic hepatitis and HCC development in the Mdr2-KO model, including inefficient anti-inflammatory activity of the endogenous lectin Gal-1 in the FVB strain.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/deficiency
- ATP Binding Cassette Transporter, Subfamily B/genetics
- Animals
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Cell Transformation, Neoplastic/pathology
- Chemical and Drug Induced Liver Injury/prevention & control
- Concanavalin A
- Galectin 1/physiology
- Hepatitis, Chronic/complications
- Hepatitis, Chronic/etiology
- Hepatitis, Chronic/pathology
- Liver/metabolism
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Male
- Methionine Adenosyltransferase/biosynthesis
- Mice
- Mice, Inbred Strains/genetics
- Mice, Knockout
- ATP-Binding Cassette Sub-Family B Member 4
Collapse
Affiliation(s)
- Tamara Potikha
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Galectin-1 (GAL-1) expression is a useful tool to differentiate between small cell osteosarcoma and Ewing sarcoma. Virchows Arch 2013; 462:665-71. [DOI: 10.1007/s00428-013-1423-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 04/07/2013] [Accepted: 05/05/2013] [Indexed: 12/28/2022]
|
40
|
Heusschen R, Griffioen AW, Thijssen VL. Galectin-9 in tumor biology: a jack of multiple trades. Biochim Biophys Acta Rev Cancer 2013; 1836:177-85. [PMID: 23648450 DOI: 10.1016/j.bbcan.2013.04.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 12/22/2022]
Abstract
Galectin family members have been shown to exert multiple roles in the context of tumor biology. Several recent findings support a similar multi-faceted role for galectin-9. Galectin-9 expression is frequently altered in cancer as compared to normal tissues. In addition, an increasing amount of evidence suggests that galectin-9 is involved in several aspects of tumor progression, including tumor cell adhesion and survival, immune escape and angiogenesis. Also, galectin-9 shows potential as a prognostic marker and a therapeutic target for several malignancies. In this review we summarize both the established and the emerging roles of galectin-9 in tumor biology and discuss the potential application of galectin-9 in anti-cancer therapy.
Collapse
Affiliation(s)
- Roy Heusschen
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
41
|
Hiramatsu H, Takeuchi K, Takeuchi H. Involvement of Histidine Residues in the pH-Dependent β-Galactoside Binding Activity of Human Galectin-1. Biochemistry 2013; 52:2371-80. [DOI: 10.1021/bi4001112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Hirotsugu Hiramatsu
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Sendai 980-8578, Japan
| | - Katsuyuki Takeuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Sendai 980-8578, Japan
| | - Hideo Takeuchi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Aobayama, Sendai 980-8578, Japan
| |
Collapse
|
42
|
|
43
|
Wu H, Chen P, Liao R, Li YW, Yi Y, Wang JX, Sun TW, Zhou J, Shi YH, Yang XR, Jin JJ, Cheng YF, Fan J, Qiu SJ. Overexpression of galectin-1 is associated with poor prognosis in human hepatocellular carcinoma following resection. J Gastroenterol Hepatol 2012; 27:1312-1319. [PMID: 22432916 DOI: 10.1111/j.1440-1746.2012.07130.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIM The high expression of the galectin-1 predicts poor patient outcome in several tumors. The aim of this study was to investigate its prognostic value in patients with hepatocellular carcinoma (HCC) after resection. METHODS Galectin-1 and tumor-infiltrating FoxP3(+) regulatory T cells (Tregs) were validated by tissue microarrays from HCC patients (n = 386) and statistically assessed for correlations with the clinical profiles and the prognosis of the patients. RESULTS We found that galectin-1, which was prevalently upregulated in HCC, was significantly associated with tumor invasive characteristics (such as vascular invasion, incomplete encapsulation, poor differentiation, multiple number, and large tumor size). Patients with high galectin-1 expression had a significantly poorer tumor recurrence (P = 0.025) and overall survival (P = 0.021) than those with low galectin-1 expression. Even in early-stage disease, high galectin-1 expression was also independently associated with shortened survival (P < 0.001) and increased tumor recurrence (P = 0.005). Multivariate Cox proportional hazards analysis showed that galectin-1 was an independent marker for predicting the poor prognosis of HCC. The galectin-1 level was positively related to the number of tumor-infiltrating FoxP3(+) Tregs (r = 0.416, P < 0.001), and their combination served as a better prognosticator. The postoperative tumor recurrence and survival of HCC patients with galectin-1(high) and FoxP3(high) were significantly poorer than the other groups (both P < 0.001). CONCLUSIONS Galectin-1 might be a new prognostic factor for HCC after resection and could potentially be a high-priority therapeutic target.
Collapse
Affiliation(s)
- Han Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Kundu JK, Surh YJ. Emerging avenues linking inflammation and cancer. Free Radic Biol Med 2012; 52:2013-37. [PMID: 22391222 DOI: 10.1016/j.freeradbiomed.2012.02.035] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 02/14/2012] [Accepted: 02/16/2012] [Indexed: 12/12/2022]
Abstract
The role of inflammation in carcinogenesis has been extensively investigated and well documented. Many biochemical processes that are altered during chronic inflammation have been implicated in tumorigenesis. These include shifting cellular redox balance toward oxidative stress; induction of genomic instability; increased DNA damage; stimulation of cell proliferation, metastasis, and angiogenesis; deregulation of cellular epigenetic control of gene expression; and inappropriate epithelial-to-mesenchymal transition. A wide array of proinflammatory cytokines, prostaglandins, nitric oxide, and matricellular proteins are closely involved in premalignant and malignant conversion of cells in a background of chronic inflammation. Inappropriate transcription of genes encoding inflammatory mediators, survival factors, and angiogenic and metastatic proteins is the key molecular event in linking inflammation and cancer. Aberrant cell signaling pathways comprising various kinases and their downstream transcription factors have been identified as the major contributors in abnormal gene expression associated with inflammation-driven carcinogenesis. The posttranscriptional regulation of gene expression by microRNAs also provides the molecular basis for linking inflammation to cancer. This review highlights the multifaceted role of inflammation in carcinogenesis in the context of altered cellular redox signaling.
Collapse
|
45
|
Peng W. Intravenous immunoglobulin treatment on anti-GM1 antibodies associated neuropathies inhibits cholera toxin and galectin-1 binding to ganglioside GM1. Immunol Lett 2012; 143:146-51. [DOI: 10.1016/j.imlet.2012.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 12/30/2011] [Accepted: 01/13/2012] [Indexed: 12/17/2022]
|