1
|
Sun R, Wang Z, Li M, Du T, Jia S, Yang W, Yang L. Regulatory Effects of Copper on Ghrelin Secretion in Rat Fundic Glands. J Anim Physiol Anim Nutr (Berl) 2025; 109:521-532. [PMID: 39545633 DOI: 10.1111/jpn.14068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Copper (Cu) is an effective additive in feed for promoting growth. Growth dan axis comprising growth hormone (GH), somatostatin (SS) and GH-releasing hormone (GHRH), with ghrelin regulating their release. The growth-promoting effects of Cu are closely related to ghrelin, but the specific mechanism behind the relationship remains unknown. We investigated the adjustment of ghrelin synthesis and secretion by Cu. Sprague-Dawley rats were fed basal diets with an addition of 0, 120 or 240 mg/kg Cu sulfate for 28 day to establish a growth-promoting model. Signalling molecules relevant to ghrelin synthesis and secretion were detected and mechanistically explored using enzyme-linked immunosorbent assay, quantitative reverse-transcription polymerase chain reaction and Western blot analysis. The 120 mg/kg supplement improved growth performance; significantly increased the serum levels of ghrelin, ghrelin O-acyltransferase (GOAT), acylated ghrelin (AG), GH, and reactive oxygen species (ROS) and decreased those of SS; significantly increased the mRNA and protein expression of ghrelin, GOAT, ghrelin receptor (GHS-R1α), and activator protein 1 (AP-1); increased the phosphorylation ratio of JNK and p38 MAPK; and inhibited the mRNA and protein expression of SS and SS receptor subtype 2 (SSTR2) in gastric fundic gland tissues. Thus, Cu may affect gastric ghrelin synthesis at the transcriptional level by activating the JNK/p38 MAPK pathway through increased ROS levels and regulating the activation of the downstream redox-sensitive transcription factor AP-1. SS plays a crucial determinant role in ghrelin regulation via intragastric Cu. Cu promotes GOAT activity and ghrelin secretion by inhibiting SS secretion, affecting AG levels, and promoting ghrelin acylation through ghrelin/GOAT/GHS-R1α system, modulating ghrelin secretion.
Collapse
Affiliation(s)
- Rui Sun
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Zhongshen Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Meng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Tianyang Du
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Shuang Jia
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Wenyan Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| | - Lianyu Yang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, Jilin Province, China
- Jilin Provincial Key Lab of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, Jilin Province, China
- Key Laboratory of Animal Production, Product Quality and Security of Ministry of Education, Changchun, Jilin Province, China
| |
Collapse
|
2
|
Ha YS, Kim TK, Heo J, Oh J, Kim SK, Kim J, Lee J, Yang SR, Hwang S, Kim SJ. Rocaglamide-A mitigates LPS-induced hepatic inflammation by modulating JNK/AP-1 signaling cascade and ROS production in hepatocytes. Toxicol Res 2025; 41:47-59. [PMID: 39802115 PMCID: PMC11717754 DOI: 10.1007/s43188-024-00263-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 01/03/2025] Open
Abstract
Lipopolysaccharide (LPS), a gut-derived endotoxin, is a recognized risk factor for both Non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). Rocaglamide-A (Roc-A), a natural compound derived from the genus Aglaia, is known for its pharmacological and immunosuppressive effects on various cell types. Although our recent investigations have unveiled Roc-A's anti-adipogenic role in adipocytes, its mechanism in hepatic inflammation remains elusive. This study delves into Roc-A's protective effects on LPS-induced hepatic inflammation. Our results demonstrated that Roc-A treatment significantly reduced the LPS-induced production of inflammatory cytokines in hepatocytes. Intriguingly, Roc-A decreased LPS-induced production of reactive oxygen species (ROS), upregulated antioxidant gene expression, and downregulated endoplasmic reticulum (ER) stress-related gene expression. Mechanistically, Roc-A significantly attenuated LPS-induced phosphorylation of c-Jun N-terminal kinase (JNK) and activator protein-1 (AP-1). Notably, this effect was abolished by the JNK activator Anisomycin, while the JNK inhibitor SP600125 enhanced it. Furthermore, Roc-A suppressed the expression of NF-κB target genes, including inducible nitric oxide synthase (iNOS), thereby alleviating iNOS-derived nitric oxide (NO) production. These findings collectively indicate that Roc-A has the potential to alleviate LPS-induced nitrosative/oxidative stress and hepatic inflammation by inhibiting JNK phosphorylation. Thus, Roc-A emerges as a promising anti-inflammatory intervention for LPS-induced hepatic inflammation. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00263-y.
Collapse
Affiliation(s)
- Yoon-su Ha
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Taek-Kyong Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Jun Heo
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Jintaek Oh
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Seung-Kyoon Kim
- Department of Convergent Bioscience and Informatics, College of Bioscience and Biotechnology, Chungnam National University, 34134 Daejeon, Republic of Korea
| | - Jeongkyu Kim
- Department of Life Science, Chung-Ang University, 06974 Seoul, Republic of Korea
| | - Jeonghyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Se-Ran Yang
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
- Department of Medicine, Kangwon National University, 24341 Chuncheon, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, 46241 Busan, Republic of Korea
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 24341 Chuncheon, Republic of Korea
- Global/Gangwon Innovative Biologics-Regional Leading Research Center (GIB-RLRC), Kangwon National University, 24341 Chuncheon, Republic of Korea
| |
Collapse
|
3
|
Kim HY, Jeong KM, Kim SH, Choi YJ, Kang HG, Jung H, Min K, Kim HM, Jeong HJ. Modulating effect of Eunkyo-san on expression of inflammatory cytokines and angiotensin-converting enzyme 2 in human mast cells. In Vitro Cell Dev Biol Anim 2024; 60:195-208. [PMID: 38228999 DOI: 10.1007/s11626-024-00847-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/19/2023] [Indexed: 01/18/2024]
Abstract
Eunkyo-san is widely used in the treatment of severe respiratory infections. Mast cells not only serve as host cells for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but also they also exacerbate Coronavirus disease in 2019 (COVID-19) by causing a cytokine storm. Here we investigated whether Eunkyo-san and its active compound naringenin regulate the expression of inflammatory cytokines and factors connected to viral infection in activated human mast cell line, HMC-1 cells. Eunkyo-san and naringenin significantly reduced levels of inflammatory cytokines including interleukin (IL)-1β, IL-6, IL-8, thymic stromal lymphopoietin, and tumor necrosis factor-α without impacting cytotoxicity. Eunkyo-san and naringenin reduced levels of factors connected to SARS-CoV-2 infection such as angiotensin-converting enzyme 2 (ACE2, SARS-CoV-2 receptor), transmembrane protease/serine subfamily member 2, and tryptase in activated HMC-1 cells. Treatment with Eunkyo-san and naringenin considerably reduced expression levels of ACE2 transcription factor, AP-1 (C-JUN and C-FOS) by blocking phosphatidylinositide-3-kinase and c-Jun NH2-terminal kinases signaling pathways. In addition, Eunkyo-san and naringenin effectively suppressed activation of signal transducer and activator of transcription 3, nuclear translocation of nuclear factor-κB, and activation of caspase-1 in activated HMC-1 cells. Furthermore, Eunkyo-san and naringenin reduced expression of ACE2 mRNA in two activated mast cell lines, RBL-2H3 and IC-2 cells. The overall study findings showed that Eunkyo-san diminished the expression levels of inflammatory cytokines and ACE2, and these findings imply that Eunkyo-san is able to effectively mitigating the cytokine storm brought on by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Biochip Research Center, Hoseo University, Asan, 31499, Republic of Korea
| | - Kyung-Min Jeong
- Division of Food and Pharmaceutical Engineering, Hoseo University, Asan, 31499, Republic of Korea
| | - Seung-Hwan Kim
- Division of Food and Pharmaceutical Engineering, Hoseo University, Asan, 31499, Republic of Korea
| | - Yu-Jin Choi
- Division of Food and Pharmaceutical Engineering, Hoseo University, Asan, 31499, Republic of Korea
| | - Ho-Geun Kang
- Department of Bio-Convergence System, Graduate School, Hoseo University, Asan, 31499, Republic of Korea
| | - Hanchul Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kyunghwon Min
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyung-Min Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan, 31499, Republic of Korea.
- Division of Food and Pharmaceutical Engineering, Hoseo University, Asan, 31499, Republic of Korea.
- Department of Bio-Convergence System, Graduate School, Hoseo University, Asan, 31499, Republic of Korea.
| |
Collapse
|
4
|
Bogut A, Stojanovic B, Jovanovic M, Dimitrijevic Stojanovic M, Gajovic N, Stojanovic BS, Balovic G, Jovanovic M, Lazovic A, Mirovic M, Jurisevic M, Jovanovic I, Mladenovic V. Galectin-1 in Pancreatic Ductal Adenocarcinoma: Bridging Tumor Biology, Immune Evasion, and Therapeutic Opportunities. Int J Mol Sci 2023; 24:15500. [PMID: 37958483 PMCID: PMC10650903 DOI: 10.3390/ijms242115500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 11/15/2023] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) remains one of the most challenging malignancies to treat, with a complex interplay of molecular pathways contributing to its aggressive nature. Galectin-1 (Gal-1), a member of the galectin family, has emerged as a pivotal player in the PDAC microenvironment, influencing various aspects from tumor growth and angiogenesis to immune modulation. This review provides a comprehensive overview of the multifaceted role of Galectin-1 in PDAC. We delve into its contributions to tumor stroma remodeling, angiogenesis, metabolic reprogramming, and potential implications for therapeutic interventions. The challenges associated with targeting Gal-1 are discussed, given its pleiotropic functions and complexities in different cellular conditions. Additionally, the promising prospects of Gal-1 inhibition, including the utilization of nanotechnology and theranostics, are highlighted. By integrating recent findings and shedding light on the intricacies of Gal-1's involvement in PDAC, this review aims to provide insights that could guide future research and therapeutic strategies.
Collapse
Affiliation(s)
- Ana Bogut
- City Medical Emergency Department, 11000 Belgrade, Serbia;
| | - Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.); (G.B.)
- Department of General Surgery, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia;
| | - Marina Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.J.); (V.M.)
| | | | - Nevena Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Bojana S. Stojanovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Goran Balovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.); (G.B.)
| | - Milan Jovanovic
- Department of Abdominal Surgery, Military Medical Academy, 11000 Belgrade, Serbia;
| | - Aleksandar Lazovic
- Department of General Surgery, University Clinical Center Kragujevac, 34000 Kragujevac, Serbia;
| | - Milos Mirovic
- Department of Surgery, General Hospital of Kotor, 85330 Kotor, Montenegro;
| | - Milena Jurisevic
- Department of Clinical Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ivan Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia;
| | - Violeta Mladenovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (M.J.); (V.M.)
| |
Collapse
|
5
|
Yu X, Qian J, Ding L, Yin S, Zhou L, Zheng S. Galectin-1: A Traditionally Immunosuppressive Protein Displays Context-Dependent Capacities. Int J Mol Sci 2023; 24:ijms24076501. [PMID: 37047471 PMCID: PMC10095249 DOI: 10.3390/ijms24076501] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
Galectin–Carbohydrate interactions are indispensable to pathogen recognition and immune response. Galectin-1, a ubiquitously expressed 14-kDa protein with an evolutionarily conserved β-galactoside binding site, translates glycoconjugate recognition into function. That galectin-1 is demonstrated to induce T cell apoptosis has led to substantial attention to the immunosuppressive properties of this protein, such as inducing naive immune cells to suppressive phenotypes, promoting recruitment of immunosuppressing cells as well as impairing functions of cytotoxic leukocytes. However, only in recent years have studies shown that galectin-1 appears to perform a pro-inflammatory role in certain diseases. In this review, we describe the anti-inflammatory function of galectin-1 and its possible mechanisms and summarize the existing therapies and preclinical efficacy relating to these agents. In the meantime, we also discuss the potential causal factors by which galectin-1 promotes the progression of inflammation.
Collapse
|
6
|
McCallion O, Bilici M, Hester J, Issa F. Regulatory T-cell therapy approaches. Clin Exp Immunol 2023; 211:96-107. [PMID: 35960852 PMCID: PMC10019137 DOI: 10.1093/cei/uxac078] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Tregs) have enormous therapeutic potential to treat a variety of immunopathologies characterized by aberrant immune activation. Adoptive transfer of ex vivo expanded autologous Tregs continues to progress through mid- to late-phase clinical trials in several disease spaces and has generated promising preliminary safety and efficacy signals to date. However, the practicalities of this strategy outside of the clinical trial setting remain challenging. Here, we review the current landscape of regulatory T-cell therapy, considering emergent approaches and technologies presenting novel ways to engage Tregs, and reflect on the progress necessary to deliver their therapeutic potential to patients.
Collapse
Affiliation(s)
- Oliver McCallion
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Merve Bilici
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Joanna Hester
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Correspondence. Fadi Issa, Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK.
| |
Collapse
|
7
|
Kim HY, Kang HG, Choi YJ, Kim HM, Jeong HJ. Caudatin attenuates inflammatory reaction by suppressing JNK/AP-1/NF-κB/caspase-1 pathways in activated HMC-1 cells. Food Sci Biotechnol 2023; 32:1101-1109. [PMID: 36683865 PMCID: PMC9844171 DOI: 10.1007/s10068-023-01251-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/21/2022] [Accepted: 01/04/2023] [Indexed: 01/18/2023] Open
Abstract
One of the interfering factors in Coronavirus disease 2019 (COVID-19) is the cytokine storm, which contributes to hyperinflammation. Mast cells cause COVID-19 hyperinflammation by increasing inflammatory cytokine levels. We investigated whether caudatin, an active compound of Cynanchum auriculatum, could suppress inflammatory response signaling in human mast cell line, HMC-1 cells. Caudatin suppressed activation of c-Jun N-terminal kinase (JNK) and activator protein-1 (AP-1) in HMC-1 cells. Caudatin suppressed nuclear translocation of catalytic subunit (p65) of nuclear factor (NF)-κB by blocking IκBα phosphorylation and degradation. Caudatin also reduced levels of activated-caspase-1 protein and activation of caspase-1. Non-toxic caudatin doses inhibited the mRNA expression and protein synthesis of pro-inflammatory cytokines. A significant finding was that caudatin inhibited JNK/AP-1/NF-κB/caspase-1 signaling molecules, reducing the secretion of inflammatory cytokines. Consequently, we propose that caudatin might be used as a material in health functional foods to alleviate mast cell-mediated inflammatory conditions like COVID-19.
Collapse
Affiliation(s)
- Hee-Yun Kim
- Biochip Research Center, Hoseo University, Asan, 31499 Republic of Korea
| | - Ho-Geun Kang
- Department of Bio-Convergence System, Graduate School, Hoseo University, Asan, 31499 Republic of Korea
| | - Yu-Jin Choi
- Department of Food Science & Technology, Hoseo University, 20 Hoseo-ro, 79 Beon-gil, Baebang-eup, Asan, 31499 Republic of Korea
| | - Hyung-Min Kim
- Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, 130-701 Republic of Korea
| | - Hyun-Ja Jeong
- Biochip Research Center, Hoseo University, Asan, 31499 Republic of Korea ,Department of Bio-Convergence System, Graduate School, Hoseo University, Asan, 31499 Republic of Korea ,Department of Food Science & Technology, Hoseo University, 20 Hoseo-ro, 79 Beon-gil, Baebang-eup, Asan, 31499 Republic of Korea
| |
Collapse
|
8
|
Lau LS, Mohammed NBB, Dimitroff CJ. Decoding Strategies to Evade Immunoregulators Galectin-1, -3, and -9 and Their Ligands as Novel Therapeutics in Cancer Immunotherapy. Int J Mol Sci 2022; 23:15554. [PMID: 36555198 PMCID: PMC9778980 DOI: 10.3390/ijms232415554] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
Galectins are a family of ß-galactoside-binding proteins that play a variety of roles in normal physiology. In cancer, their expression levels are typically elevated and often associated with poor prognosis. They are known to fuel a variety of cancer progression pathways through their glycan-binding interactions with cancer, stromal, and immune cell surfaces. Of the 15 galectins in mammals, galectin (Gal)-1, -3, and -9 are particularly notable for their critical roles in tumor immune escape. While these galectins play integral roles in promoting cancer progression, they are also instrumental in regulating the survival, differentiation, and function of anti-tumor T cells that compromise anti-tumor immunity and weaken novel immunotherapies. To this end, there has been a surge in the development of new strategies to inhibit their pro-malignancy characteristics, particularly in reversing tumor immunosuppression through galectin-glycan ligand-targeting methods. This review examines some new approaches to evading Gal-1, -3, and -9-ligand interactions to interfere with their tumor-promoting and immunoregulating activities. Whether using neutralizing antibodies, synthetic peptides, glyco-metabolic modifiers, competitive inhibitors, vaccines, gene editing, exo-glycan modification, or chimeric antigen receptor (CAR)-T cells, these methods offer new hope of synergizing their inhibitory effects with current immunotherapeutic methods and yielding highly effective, durable responses.
Collapse
Affiliation(s)
- Lee Seng Lau
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Norhan B. B. Mohammed
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
- Department of Medical Biochemistry, Faculty of Medicine, South Valley University, Qena 83523, Egypt
| | - Charles J. Dimitroff
- Department of Translational Medicine, Translational Glycobiology Institute at FIU, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
9
|
Jiang Z, Zhang W, Sha G, Wang D, Tang D. Galectins Are Central Mediators of Immune Escape in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:5475. [PMID: 36428567 PMCID: PMC9688059 DOI: 10.3390/cancers14225475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 11/09/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers and is highly immune tolerant. Although there is immune cell infiltration in PDAC tissues, most of the immune cells do not function properly and, therefore, the prognosis of PDAC is very poor. Galectins are carbohydrate-binding proteins that are intimately involved in the proliferation and metastasis of tumor cells and, in particular, play a crucial role in the immune evasion of tumor cells. Galectins induce abnormal functions and reduce numbers of tumor-associated macrophages (TAM), natural killer cells (NK), T cells and B cells. It further promotes fibrosis of tissues surrounding PDAC, enhances local cellular metabolism, and ultimately constructs tumor immune privileged areas to induce immune evasion behavior of tumor cells. Here, we summarize the respective mechanisms of action played by different Galectins in the process of immune escape from PDAC, focusing on the mechanism of action of Galectin-1. Galectins cause imbalance between tumor immunity and anti-tumor immunity by coordinating the function and number of immune cells, which leads to the development and progression of PDAC.
Collapse
Affiliation(s)
- Zhengting Jiang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Gengyu Sha
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
| | - Daorong Wang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225000, China
| | - Dong Tang
- Clinical Medical College, Yangzhou University, Yangzhou 225000, China
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Northern Jiangsu People’s Hospital, Yangzhou 225000, China
| |
Collapse
|
10
|
Immunoregulatory signal networks and tumor immune evasion mechanisms: insights into therapeutic targets and agents in clinical development. Biochem J 2022; 479:2219-2260. [DOI: 10.1042/bcj20210233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
Through activation of immune cells, the immune system is responsible for identifying and destroying infected or otherwise damaged cells including tumorigenic cells that can be recognized as foreign, thus maintaining homeostasis. However, tumor cells have evolved several mechanisms to avoid immune cell detection and killing, resulting in tumor growth and progression. In the tumor microenvironment, tumor infiltrating immune cells are inactivated by soluble factors or tumor promoting conditions and lose their effects on tumor cells. Analysis of signaling and crosstalk between immune cells and tumor cells have helped us to understand in more detail the mechanisms of tumor immune evasion and this forms basis for drug development strategies in the area of cancer immunotherapy. In this review, we will summarize the dominant signaling networks involved in immune escape and describe the status of development of therapeutic strategies to target tumor immune evasion mechanisms with focus on how the tumor microenvironment interacts with T cells.
Collapse
|
11
|
Biswas S, Tikader B, Kar S, Viswanathan GA. Modulation of signaling cross-talk between pJNK and pAKT generates optimal apoptotic response. PLoS Comput Biol 2022; 18:e1010626. [PMID: 36240239 PMCID: PMC9604984 DOI: 10.1371/journal.pcbi.1010626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 10/26/2022] [Accepted: 10/03/2022] [Indexed: 01/25/2023] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a well-known modulator of apoptosis by maintaining a balance between proliferation and cell-death in normal cells. Cancer cells often evade apoptotic response following TNFα stimulation by altering signaling cross-talks. Thus, varying the extent of signaling cross-talk could enable optimal TNFα mediated apoptotic dynamics. Herein, we use an experimental data-driven mathematical modeling to quantitate the extent of synergistic signaling cross-talk between the intracellular entities phosphorylated JNK (pJNK) and phosphorylated AKT (pAKT) that orchestrate the phenotypic apoptosis level by modulating the activated Caspase3 dynamics. Our study reveals that this modulation is orchestrated by the distinct dynamic nature of the synergism at early and late phases. We show that this synergism in signal flow is governed by branches originating from either TNFα receptor and NFκB, which facilitates signaling through survival pathways. We demonstrate that the experimentally quantified apoptosis levels semi-quantitatively correlates with the model simulated Caspase3 transients. Interestingly, perturbing pJNK and pAKT transient dynamics fine-tunes this accumulated Caspase3 guided apoptotic response. Thus, our study offers useful insights for identifying potential targeted therapies for optimal apoptotic response.
Collapse
Affiliation(s)
- Sharmila Biswas
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
| | - Baishakhi Tikader
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
| | - Sandip Kar
- Department of Chemistry, Indian Institute of Technology Bombay, Mumbai, India
- * E-mail: (SK); (GAV)
| | - Ganesh A. Viswanathan
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, India
- * E-mail: (SK); (GAV)
| |
Collapse
|
12
|
Zhang Y, Li Q, Jiang N, Su Z, Yuan Q, Lv L, Sang X, Chen R, Feng Y, Chen Q. Dihydroartemisinin beneficially regulates splenic immune cell heterogeneity through the SOD3-JNK-AP-1 axis. SCIENCE CHINA. LIFE SCIENCES 2022; 65:1636-1654. [PMID: 35226255 DOI: 10.1007/s11427-021-2061-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022]
Abstract
The immunomodulatory potential of dihydroartemisinin (DHA) has recently been highlighted; however, the potential mechanism remains to be clarified. Single-cell RNA sequencing was explored in combination with cellular and biochemical approaches to elucidate the immunomodulatory mechanisms of DHA. In this study, we found that DHA induced both spleen enlargement and rearrangement of splenic immune cell subsets in mice. It was revealed that DHA promoted the reversible expansion of effective regulatory T cells and interferon-γ+ cytotoxic CD8+ T cells in the spleen via induction of superoxide dismutase 3 (SOD3) expression and increased phosphorylation of c-Jun N-terminal kinases (JNK) and its downstream activator protein 1 (AP-1) transcription factors. Further, SOD3 knockout mice were resistant to the regulatory effect of DHA. Thus, DHA, through the activation of the SOD3-JNK-AP-1 axis, beneficially regulated immune cell heterogeneity and splenic immune cell homeostasis to treat autoimmune diseases.
Collapse
Affiliation(s)
- Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Qilong Li
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ziwei Su
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Quan Yuan
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Lei Lv
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.
- Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, Shenyang, 110866, China.
| |
Collapse
|
13
|
Galectin-1-Dependent Mitochondria Apoptosis Plays an Essential Role in the Potential Protein Targets of DBDCT-Induced Hepatotoxicity as Revealed by Quantitative Proteomic Analyses. Bioinorg Chem Appl 2022; 2022:5176300. [PMID: 35154294 PMCID: PMC8824758 DOI: 10.1155/2022/5176300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/31/2021] [Indexed: 11/20/2022] Open
Abstract
Di-n-butyl-di-(4-chlorobenzohydroxamato) tin(IV) (DBDCT), a new patent agent, exhibited strong antitumor activity. In some cases, its activity was close to or even higher than cisplatin, a first-line clinical metallic agent. Similar to platinum compounds, it also showed toxicity. However, the effective targets and mechanisms for specific toxicity and biological activity are still unclear. In this study, proteomic analysis revealed that 146 proteins (98 upregulated and 48 downregulated) were differentially identified by label-free LC-MS/MS after DBDCT treatment. Meanwhile, network analysis of these differential proteins suggested that protein Galectin-1 (Gal-1) could regulate the apoptosis process (15 related proteins), which played an essential role in the potential targets of DBDCT-induced hepatotoxicity. Furthermore, it was demonstrated that DBDCT might promote ROS production, activate NF-κB p65, inhibit Ras and p-ERK1/2 expressions, increase the level of Gal-1, subsequently upregulate the expressions of Bax, p53, Fas, and FasL, and downregulate the expression of Bcl-2. As a result of these modulations, caspase cascades were finally activated, which executed apoptosis in HL7702 liver cells. Correspondingly, NAC (inhibitor of ROS), PDTC (inhibitor of NF-κB), EGF (ERK1/2 activator), and OTX008 (inhibitor of Gal-1) were found to reverse and abolish the DBDCT-associated cytotoxicity partially. In conclusion, Gal-1 might be the potential target for toxicity and biological activity. Moreover, the present study will lay the groundwork for future research about di-n-butyl-di-(4-chlorobenzohydroxamato) tin structure optimization and developing it into a new potential anticancer agent.
Collapse
|
14
|
Ye F, Dan G, Zhao Y, Yu W, Cheng J, Chen M, Sai Y, Zou Z. Small-interfering RNA for c-Jun attenuates cell death by preventing JNK-dependent PARP1 cleavage and DNA fragmentation in nitrogen mustard-injured immortalized human bronchial epithelial cells. Toxicol Res (Camb) 2021; 10:1034-1044. [PMID: 34733488 DOI: 10.1093/toxres/tfab081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/29/2021] [Accepted: 07/27/2021] [Indexed: 11/14/2022] Open
Abstract
Sulfur mustard (a type of vesicant) can directly damage lung bronchial epithelium via aerosol inhalation, and prevalent cell death is an early event that obstructs the respiratory tract. JNK/c-Jun is a stress response pathway, but its role in cell death of the injured cells is not clear. Here, we report that JNK/c-Jun was activated in immortalized human bronchial epithelial (HBE) cells exposed to a lethal dose (20 μM) of nitrogen mustard (NM, a sulfur mustard analog). c-Jun silencing using small-interfering RNA (siRNA) rendered the cells resistant to NM-mediated cell death by blocking poly(ADP-ribose) polymerase 1 (PARP1) cleavage and DNA fragmentation. In addition, the transduction of upstream extrinsic (Fasl-Fas-caspase-8) and intrinsic (loss of Bcl-2 and mitochondrial membrane potential, ΔΨm) apoptosis pathways, as well as phosphorylated (p)-H2AX (Ser139), an epigenetic marker contributing to DNA fragmentation and PARP1 activity, was partially suppressed. To mimic the detachment of cells by NM, HBE cells were trypsinized and seeded on culture plates that were pre-coated with poly-HEMA to prevent cell adhesion. The JNK/c-Jun pathway was found to be activated in the detached cells. In conclusion, our results indicate that JNK/c-Jun pathway activation is necessary for NM-caused HBE cell death and further suggest that c-Jun silencing may be a potential approach to protect HBE cells from vesicant damage.
Collapse
Affiliation(s)
- Feng Ye
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Guorong Dan
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yuanpeng Zhao
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wenpei Yu
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jin Cheng
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Mingliang Chen
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yan Sai
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhongmin Zou
- Department of Chemical Defense Medicine, School of Military Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| |
Collapse
|
15
|
Bhalla M, Heinzinger LR, Morenikeji OB, Marzullo B, Thomas BN, Bou Ghanem EN. Transcriptome Profiling Reveals CD73 and Age-Driven Changes in Neutrophil Responses against Streptococcus pneumoniae. Infect Immun 2021; 89:e0025821. [PMID: 34310891 PMCID: PMC8519284 DOI: 10.1128/iai.00258-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/15/2021] [Indexed: 11/20/2022] Open
Abstract
Neutrophils are required for host resistance against Streptococcus pneumoniae, but their function declines with age. We previously found that CD73, an enzyme required for antimicrobial activity, is downregulated in neutrophils (also known as polymorphonuclear leukocytes [PMNs]) from aged mice. This study explored transcriptional changes in neutrophils induced by S. pneumoniae to identify pathways controlled by CD73 and dysregulated with age. Pure bone marrow-derived neutrophils isolated from wild-type (WT) young and old and CD73 knockout (CD73KO) young mice were mock challenged or infected with S. pneumoniae ex vivo. RNA sequencing (RNA-Seq) was performed to identify differentially expressed genes (DEGs). We found that infection triggered distinct global transcriptional changes across hosts that were strongest in CD73KO neutrophils. Surprisingly, there were more downregulated than upregulated genes in all groups upon infection. Downregulated DEGs indicated a dampening of immune responses in old and CD73KO hosts. Further analysis revealed that CD73KO neutrophils expressed higher numbers of long noncoding RNAs (lncRNAs) than those in WT controls. Predicted network analysis indicated that CD73KO-specific lncRNAs control several signaling pathways. We found that genes in the c-Jun N-terminal kinase (JNK)-mitogen-activated protein kinase (MAPK) pathway were upregulated upon infection in CD73KO mice and in WT old mice, but not in WT young mice. This corresponded to functional differences, as phosphorylation of the downstream AP-1 transcription factor component c-Jun was significantly higher in neutrophils from infected CD73KO mice and old mice. Importantly, inhibition of JNK/AP-1 rescued the ability of these neutrophils to kill S. pneumoniae. Together, our findings revealed that the ability of neutrophils to modify their gene expression to better adapt to bacterial infection is in part regulated by CD73 and declines with age.
Collapse
Affiliation(s)
- Manmeet Bhalla
- Department of Microbiology and Immunology, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| | - Lauren R. Heinzinger
- Department of Microbiology and Immunology, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| | - Olanrewaju B. Morenikeji
- Department of Biomedical Sciences, College of Health Sciences and Technology, Rochester Institute of Technology, Rochester, New York, USA
- Division of Biological and Health Sciences, University of Pittsburgh–Bradford, Bradford, Pennsylvania, USA
| | - Brandon Marzullo
- Department of Biochemistry and Center of Excellence in Bioinformatics and Life Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Bolaji N. Thomas
- Department of Biomedical Sciences, College of Health Sciences and Technology, Rochester Institute of Technology, Rochester, New York, USA
| | - Elsa N. Bou Ghanem
- Department of Microbiology and Immunology, State University of New York at Buffalo School of Medicine, Buffalo, New York, USA
| |
Collapse
|
16
|
Zou J, Yi S, Niu L, Zhou H, Lin Z, Wang Y, Huang X, Meng W, Guo Y, Qi L, Meng L. Neuroprotective Effect of Ultrasound Neuromodulation on Kainic Acid- Induced Epilepsy in Mice. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:3006-3016. [PMID: 33979280 DOI: 10.1109/tuffc.2021.3079628] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Preliminary evidence suggests that low-intensity pulsed ultrasound (LIPUS) has neuroprotective effects on ischemic stroke, depression, and other conditions leading to neuronal cell death (e.g., Parkinson's disease). The purpose of this study was to investigate the neuroprotective effects of LIPUS in epileptic mice. Mice were made epileptic through kainic acid (KA) administration and then stimulated with LIPUS. The neuroprotective effect of ultrasound was evaluated by observing the latency, anxiety-like behavior, and levels of proteins related to inflammation, apoptosis, or signaling pathways. The safety of LIPUS was assessed by hematoxylin and eosin (H&E) and Nissl stainings. LIPUS prolonged the latency (Sham: 6.00 ± 0.26 days; 1-kHz pulse repetition frequency (PRF): 7.00 ± 0.31 days), improved the anxiety-like behavior, and inhibited the expression of inflammatory factors and apoptosis-related proteins. In addition, H&E and Nissl staining results confirmed that LIPUS did not damage the brain. These findings suggest that LIPUS has neuroprotective effects in mice with KA-induced epilepsy. LIPUS may offer a new therapeutic approach to epilepsy.
Collapse
|
17
|
Lymphoid Organ Proteomes Identify Therapeutic Efficacy Biomarkers Following the Intracavitary Administration of Curcumin in a Highly Invasive Rat Model of Peritoneal Mesothelioma. Int J Mol Sci 2021; 22:ijms22168566. [PMID: 34445271 PMCID: PMC8395293 DOI: 10.3390/ijms22168566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/16/2022] Open
Abstract
This study aimed to identify the proteomic changes produced by curcumin treatment following stimulation of the host immune system in a rat model of malignant mesothelioma. We analyzed the proteomes of secondary lymphoid organs from four normal rats, four untreated tumor-bearing rats, and four tumor-bearing rats receiving repeated intraperitoneal administrations of curcumin. Cross-comparing proteome analyses of histological sections of the spleen from the three groups first identified a list of eighty-three biomarkers of interest, thirteen of which corresponded to proteins already reported in the literature and involved in the anticancer therapeutic effects of curcumin. In a second step, comparing these data with proteomic analyses of histological sections of mesenteric lymph nodes revealed eight common biomarkers showing a similar pattern of changes in both lymphoid organs. Additional findings included a partial reduction of the increase in spleen-circulating biomarkers, a decrease in C-reactive protein and complement C3 in the spleen and lymph nodes, and an increase in lymph node purine nucleoside phosphorylase previously associated with liver immunodeficiency. Our results suggest some protein abundance changes could be related to the systemic, distant non-target antitumor effects produced by this phytochemical.
Collapse
|
18
|
Galectin-1 Expression Is Associated with the Response and Survival Following Preoperative Chemoradiotherapy in Locally Advanced Esophageal Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13133147. [PMID: 34201887 PMCID: PMC8268777 DOI: 10.3390/cancers13133147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/29/2023] Open
Abstract
Simple Summary Galectin-1 has been found to be involved in therapeutic resistance in a variety of cancers. However, the prognostic significance of galectin-1 expression in patients with locally advanced esophageal squamous cell carcinoma (ESCC) treated with chemoradiotherapy remains unknown. Immunohistochemically, we observed that galectin-1 overexpression in pretreatment biopsied specimens was significantly associated with a lower pathological complete response rate, worse overall survival and disease-free survival in 93 patients with locally advanced ESCC receiving preoperative chemoradiotherapy. Our findings suggest that galectin-1 may be a potential therapeutic target for patients with ESCC treated with preoperative chemoradiotherapy. Abstract The galectin-1 has been found to be involved in poor outcomes after treatment of a variety of cancers. To the best of our knowledge, however, the significance of galectin-1 expression in the sensitivity to chemoradiotherapy (CCRT) of patients with locally advanced esophageal squamous cell carcinoma (ESCC) remains unclear. Expression levels of galectin-1 were evaluated by immunohistochemistry and correlated with the treatment outcome in 93 patients with locally advanced ESCC who received preoperative CCRT between 1999 and 2012. Galectin-1 expression was significantly associated with the pathological complete response (pCR). The pCR rates were 36.1% and 13.0% (p = 0.01) in patients with low and high galectin-1 expression, respectively. Univariate analyses revealed that galectin-1 overexpression, clinical 7th American Joint Committee on Cancer (AJCC) stage III and a positive surgical margin were significant factors of worse overall survival and disease-free survival. In multivariate analyses, galectin-1 overexpression and a positive surgical margin represented the independent adverse prognosticators. Therefore, galectin-1 expression both affects the pCR and survival in patients with locally advanced ESCC receiving preoperative CCRT. Our results suggest that galectin-1 may be a potentially therapeutic target for patients with ESCC treated with preoperative CCRT.
Collapse
|
19
|
Xu WD, Huang Q, Huang AF. Emerging role of galectin family in inflammatory autoimmune diseases. Autoimmun Rev 2021; 20:102847. [PMID: 33971347 DOI: 10.1016/j.autrev.2021.102847] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/12/2021] [Accepted: 03/20/2021] [Indexed: 12/13/2022]
Abstract
Galectin family is a group of glycan-binding proteins. Members in this family are expressed in different tissues, immune or non-immune cells. These molecules are important regulators in innate and adaptive immune response, performing significantly in a broad range of cellular and pathophysiological functions, such as cell proliferation, adhesion, migration, and invasion. Findings have shown that expression of galectins is abnormal in many inflammatory autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, osteoarthritis, sjögren's syndrome, systemic sclerosis. Galectins also function as intracellular and extracellular disease regulators mainly through the binding of their carbohydrate recognition domain to glycoconjugates. Here, we review the state-of-the-art of the role that different galectin family members play in immune cells, contributing to the complex inflammatory diseases. Hopefully collection of the information will provide a preliminary theoretical basis for the exploration of new targets for treatment of the disorders.
Collapse
Affiliation(s)
- Wang-Dong Xu
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China.
| | - Qi Huang
- Department of Evidence-Based Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Fang Huang
- Department of Rheumatology and Immunology, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
20
|
Liu B, Hou Q, Ma Y, Han X. HIPK3 Mediates Inflammatory Cytokines and Oxidative Stress Markers in Monocytes in a Rat Model of Sepsis Through the JNK/c-Jun Signaling Pathway. Inflammation 2021; 43:1127-1142. [PMID: 32356246 DOI: 10.1007/s10753-020-01200-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sepsis is a fetal immunological disorder and its complication worsens in the patients with hemodialysis which may increase the risk of death. In the present study, we aimed to investigate the effect of homeodomain-interacting protein kinase 3 (HIPK3) on inflammatory factors and oxidative stress markers in monocytes of rats with sepsis by regulating the c-Jun amino-terminal kinase (JNK)/c-Jun signaling pathway. A rat model of sepsis was initially established using cecal ligation and puncture (CLP) and was further identified by enlarged spleen tissues, inflammation, and oxidative stress. Monocytes were isolated from rats with CLP-induced sepsis. HIPK3 was observed to be downregulated while JUN was upregulated in monocytes from rats with CLP-induced sepsis. Furthermore, isolated monocytes were transduced with lentiviral vectors expressing HIPK3 or shRNA against HIPK3 to explore the effect of HIPK3 on viability and apoptosis of monocytes as well as inflammatory factors and oxidative stress markers. The obtained data exhibited that overexpression of HIPK3 or inhibition of the JNK signaling pathway enhanced proliferation, reduced apoptosis of monocytes, alleviated inflammation, and oxidative stress injury. Consistently, our results may provide evidence that HIPK3 could inhibit the JNK/c-Jun signaling pathway, thereby potentially retarding the progression of sepsis.
Collapse
Affiliation(s)
- Ben Liu
- Department of Clinical Laboratory, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, No. 1 West Yellow River Road, Huai'an, 223300, Jiangsu, People's Republic of China
| | - Qiuyue Hou
- Department of Clinical Laboratory, Huaiyin Hospital of Huai'an City, 38 Beijing West Road, Huai'an, 223300, Jiangsu, People's Republic of China
| | - Yuhong Ma
- Department of Psychiatry, Huaian No. 3 People's Hospital, 272 Huaihai West Road, Huai'an, 223300, Jiangsu, People's Republic of China
| | - Xuehua Han
- Department of Clinical Laboratory, Huai'an Hospital Affiliated of Xuzhou Medical University, No. 62, Huaihai South Road, Huai'an, 223002, Jiangsu Province, People's Republic of China.
| |
Collapse
|
21
|
Ghosh DD, Lee D, Jin X, Horvitz HR, Nitabach MN. C. elegans discriminates colors to guide foraging. Science 2021; 371:1059-1063. [PMID: 33674494 PMCID: PMC8554940 DOI: 10.1126/science.abd3010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 01/08/2021] [Indexed: 12/14/2022]
Abstract
Color detection is used by animals of diverse phyla to navigate colorful natural environments and is thought to require evolutionarily conserved opsin photoreceptor genes. We report that Caenorhabditis elegans roundworms can discriminate between colors despite the fact that they lack eyes and opsins. Specifically, we found that white light guides C. elegans foraging decisions away from a blue-pigment toxin secreted by harmful bacteria. These foraging decisions are guided by specific blue-to-amber ratios of light. The color specificity of color-dependent foraging varies notably among wild C. elegans strains, which indicates that color discrimination is ecologically important. We identified two evolutionarily conserved cellular stress response genes required for opsin-independent, color-dependent foraging by C. elegans, and we speculate that cellular stress response pathways can mediate spectral discrimination by photosensitive cells and organisms-even by those lacking opsins.
Collapse
Affiliation(s)
- D Dipon Ghosh
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA.
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dongyeop Lee
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Jin
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - H Robert Horvitz
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael N Nitabach
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA.
- Department of Genetics, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| |
Collapse
|
22
|
Farhadi SA, Fettis MM, Liu R, Hudalla GA. A Synthetic Tetramer of Galectin-1 and Galectin-3 Amplifies Pro-apoptotic Signaling by Integrating the Activity of Both Galectins. Front Chem 2020; 7:898. [PMID: 31998689 PMCID: PMC6966408 DOI: 10.3389/fchem.2019.00898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/12/2019] [Indexed: 01/16/2023] Open
Abstract
Galectin-1 (G1) and galectin-3 (G3) are carbohydrate-binding proteins that can signal apoptosis in T cells. We recently reported that a synthetic tetramer with two G1 and two G3 domains ("G1/G3 Zipper") induces Jurkat T cell death more potently than G1. The pro-apoptotic signaling pathway of G1/G3 Zipper was not elucidated, but we hypothesized based on prior work that the G1 domains acted as the signaling units, while the G3 domains served as anchors that increase glycan-binding affinity. To test this, here we studied the involvement of different cell membrane glycoproteins and intracellular mediators in pro-apoptotic signaling via G1/G3 Zipper, G1, and G3. G1/G3 Zipper induced Jurkat T cell death more potently than G1 and G3 alone or in combination. G1/G3 Zipper, G1, and G3 increased caspase-8 activity, yet only G1 and G3 depended on it to induce cell death. G3 increased caspase-3 activity more than G1/G3 Zipper and G1, while all three galectin variants required it to induce cell death. JNK activation had similar roles downstream of G1/G3 Zipper, G1, and G3, whereas ERK had differing roles. CD45 was essential for G1 activity, and was involved in signaling via G1/G3 Zipper and G3. CD7 inhibited G1/G3 Zipper activity at low galectin concentrations but not at high galectin concentrations. In contrast, CD7 was necessary for G1 and G3 signaling at low galectin concentration but antagonistic at high galectin concentrations. Collectively, these observations suggest that G1/G3 Zipper amplifies pro-apoptotic signaling through the integrated activity of both the G1 and G3 domains.
Collapse
Affiliation(s)
- Shaheen A Farhadi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Margaret M Fettis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Renjie Liu
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Gregory A Hudalla
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| |
Collapse
|
23
|
Zhao T, Chen H, Cheng C, Zhang J, Yan Z, Kuang J, Kong F, Li C, Lu Q. Liraglutide protects high-glucose-stimulated fibroblasts by activating the CD36-JNK-AP1 pathway to downregulate P4HA1. Biomed Pharmacother 2019; 118:109224. [PMID: 31349139 DOI: 10.1016/j.biopha.2019.109224] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 07/10/2019] [Accepted: 07/10/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a serious complication of diabetes mellitus. It's known that glucagon-like peptide-1 (GLP-1) and prolyl 4-hydroxylase subunit alpha-1 (P4HA1) have significant effect on cardiovascular function, but their interaction in cardiac fibroblasts (CFs) is still being unraveled. METHODS AND RESULTS The present study demonstrated that glucose promotes CFs proliferation and cardiac fibrosis. Using qRT-PCR, Western blot, CCK-8, EdU, flow cytometry, wound healing and Transwell assays to explore the functions of liraglutide and P4HA1 in high-glucose (HG)-induced CFs, we proved that liraglutide as well as silencing of P4HA1 inhibited cell proliferation, migration and invasion, and promoted cell cycle arrest and apoptosis in HG-induced CFs. In addition, liraglutide downregulated P4HA1 expression, upregulated CD36 and P-JNK expression levels, and enhanced the DNA binding activity of AP-1 on P4HA1. Inhibition of CD36 or p--JNK promoted P4HA1 expression. CONCLUSIONS Liraglutide may down-regulate P4HA1 expression at least partly though CD36-JNK-AP1 pathway, thereby reducing myocardial fibrosis. Therefore, our study provides novel insight into the molecular mechanism and function of liraglutide in HG-mediated CFs.
Collapse
Affiliation(s)
- Tong Zhao
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Huiqiang Chen
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Chao Cheng
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Juan Zhang
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Zhi Yan
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Jiangying Kuang
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Feng Kong
- Central Research Laboratory, The Second Hospital of Shandong University, Jinan 250033, Shandong, China
| | - Chunyan Li
- Department of Gynaecology, Provincial Hospital Affiliated to Shandong University, Jinan 250021, Shandong, China.
| | - Qinghua Lu
- Department of Cardiology, The Second Hospital of Shandong University, Jinan 250033, Shandong, China.
| |
Collapse
|
24
|
Liu C, Li H, Zheng H, Zhai M, Lu F, Dong S, Fang T, Zhang W. CaSR activates PKCδ to induce cardiomyocyte apoptosis via ER stress‑associated apoptotic pathways during ischemia/reperfusion. Int J Mol Med 2019; 44:1117-1126. [PMID: 31257458 DOI: 10.3892/ijmm.2019.4255] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/17/2019] [Indexed: 11/06/2022] Open
Abstract
Endoplasmic reticulum (ER) stress can be activated by ischemia/reperfusion (I/R) injury in cardiomyocytes. Persistent ER stress, with an increase in intracellular Ca2+ ([Ca2+]i) concentration, leads to apoptosis. Protein kinase C (PKC) has a key role in myocardial damage by elevation of [Ca2+]i. The calcium‑sensing receptor (CaSR), a G protein‑coupled receptor, can increase the release of [Ca2+]i from the ER through the inositol triphosphate receptor (IP3R). Intracellular calcium overload has been demonstrated to cause cardiac myocyte apoptosis during I/R. However, the associations between PKC, CaSR and ER stress are not clear. The present study examined the hypothesis that activation of PKCδ by CaSR participates in ER stress‑associated apoptotic pathways within myocardial I/R. Rat hearts were subjected to 30 min of ischemia in vivo, followed by reperfusion for 120 min. GdCl3 (a CaSR activator) was used to elevate the intracellular Ca2+ concentration, but the Ca2+ concentration in the ER was significantly decreased during I/R. Following exposure to GdCl3, expression levels of CaSR, glucose‑regulated protein 78 (GRP78), Caspase‑12, phosphorylated JNK and Caspase‑3 were increased, and the ratios of apoptotic myocardial cells were significantly increased. By contrast, following exposure to rottlerin, a PKCδ inhibitor, the expression levels of these proteins and the ratio of apoptotic myocardial cells were significantly reduced. The present study also demonstrated that PKCδ translocated into the ER to induce an ER stress response and participate in the ER stress‑related apoptosis pathway. These results confirmed that CaSR activated PKCδ to induce cardiomyocyte apoptosis through ER stress‑associated apoptotic pathways during I/R in vivo.
Collapse
Affiliation(s)
- Chong Liu
- Department of Anesthesiology, Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin 300140, P.R. China
| | - Huanming Li
- Department of Cardiology, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, Tianjin 300140, P.R. China
| | - Huishuang Zheng
- Department of Pathology, Haiyang Renmin Hospital, Haiyang, Shandong 265100, P.R. China
| | - Meili Zhai
- Department of Anesthesiology, Tianjin Central Hospital of Gynecology Obstetrics, Central Gynecology Obstetrics Hospital of Nankai University, Tianjin 300052, P.R. China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Tao Fang
- Central Laboratory, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, Tianjin 300140, P.R. China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
25
|
Activator protein-1 and caspase 8 mediate p38α MAPK-dependent cardiomyocyte apoptosis induced by palmitic acid. Apoptosis 2019; 24:395-403. [DOI: 10.1007/s10495-018-01510-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
26
|
You JL, Wang W, Tang MY, Ye YH, Liu AX, Zhu YM. A potential role of galectin-1 in promoting mouse trophoblast stem cell differentiation. Mol Cell Endocrinol 2018; 470:228-239. [PMID: 29122660 DOI: 10.1016/j.mce.2017.11.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 11/03/2017] [Accepted: 11/03/2017] [Indexed: 11/24/2022]
Abstract
Galectin-1 is highly expressed in blastocysts and trophoblast giant cells during implantation, and dysregulated galectin-1 is associated with many pregnancy-related abnormalities. Elevated galectin-1 contributes to cancer cells invasion. Here, we found that galectin-1 is expressed in mouse oocytes, preimplantation embryos (all stages), and trophoblast stem (TS) cells. Peak levels of galectin-1 mRNA and protein were detected on day 4 and day 5 after the induction of TS cells differentiation. Overexpression of galectin-1 increased TS cells migration and invasion, whereas knockdown of galectin-1 attenuated these effects. Additionally, knockdown of galectin-1 in TS cells decreased the expression of matrix metalloproteinase (MMP) 2/9, ZEB-1, Snail, N-cadherin, TGF-β, Nodal, and phospho-Smad2/3, whereas the expression of E-cadherin was increased. In contrast, overexpression of galectin-1 in TS cells increased the expression of MMP2/9, ZEB-1, and N-cadherin, whereas the expression of E-cadherin was decreased. These findings suggest a potential role of galectin-1 in the differentiation of mouse TS cells.
Collapse
Affiliation(s)
- Jia-Li You
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min-Yue Tang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ying-Hui Ye
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ai-Xia Liu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Yi-Min Zhu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
Dai W, Miller WP, Toro AL, Black AJ, Dierschke SK, Feehan RP, Kimball SR, Dennis MD. Deletion of the stress-response protein REDD1 promotes ceramide-induced retinal cell death and JNK activation. FASEB J 2018; 32:fj201800413RR. [PMID: 29920218 PMCID: PMC6219834 DOI: 10.1096/fj.201800413rr] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of dyslipidemia in the development of retinal dysfunction remains poorly understood. Using an animal model of diet-induced obesity/pre-type 2 diabetes, we investigated molecular defects in the retina arising from consumption of a diet high in saturated fats and sugars ( i.e., a Western diet). We found that feeding mice a Western diet increased the abundance of retinal sphingolipids, attenuated protein kinase B (Akt) phosphorylation, enhanced JNK activation, and increased retinal cell death. When we used palmitate or C6-ceramide (Cer) to assess sphingolipid-mediated signaling in cultured murine and human cells, we observed similar effects on Akt, JNK, and cell death. Furthermore, both Western diet and C6-Cer exposure enhanced expression of the stress-response protein regulated in development and DNA damage response 1 (REDD1) and loss of REDD1 increased C6-Cer-induced JNK activation and cell death. Exogenous REDD1 expression repressed JNK-mediated phosphorylation in cultured cells. We found that thioredoxin-interacting protein (TXNIP) expression was elevated in REDD1-deficient cell lines and C6-Cer promoted TXNIP expression in both wild-type and REDD1-deficient cells. Likewise, TXNIP knockdown attenuated JNK activation and caspase 3 cleavage after either C6-Cer exposure or REDD1 deletion. The results support a model wherein Cer-induced REDD1 expression attenuates TXNIP-dependent JNK activation and retinal cell death.-Dai, W., Miller, W. P., Toro, A. L., Black, A. J., Dierschke, S. K., Feehan, R. P., Kimball, S. R., Dennis, M. D. Deletion of the stress-response protein REDD1 promotes ceramide-induced retinal cell death and JNK activation.
Collapse
Affiliation(s)
- Weiwei Dai
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - William P Miller
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Allyson L Toro
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Adam J Black
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Sadie K Dierschke
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Scot R Kimball
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Michael D Dennis
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
28
|
The N-terminal tail coordinates with carbohydrate recognition domain to mediate galectin-3 induced apoptosis in T cells. Oncotarget 2018; 8:49824-49838. [PMID: 28548942 PMCID: PMC5564810 DOI: 10.18632/oncotarget.17760] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
Galectin-3 is a galectin with a unique flexible N-terminal tail (NT) connected to the conserved carbohydrate recognition domain (CRD). Galectin-3 is associated with tumor immune tolerance and exhibits an ability to induce T cell apoptosis. We used Jurkat, Jurkat E6-1 and CEM T-cell lines and human peripheral blood mononuclear cells (PBMCs) to investigate the specific roles of the CRD and NT in inducing T cell apoptosis. Galectin-3 triggered sustained extracellular signal-regulated kinase (ERK) phosphorylation that induced apoptosis. ERK was situated upstream of caspase-9 and was independently activated by reactive oxygen species (ROS) and protein kinase C (PKC). The first twelve NT residues had no role in the apoptosis. Residues 13-68 were essential for activating ROS, but did not activate PKC. However, residues 69-110 were required for activation of PKC. An NT fragment and a NT-specific antibody antagonized the apoptosis triggered by full-length galectin-3 further supporting our findings. These findings indicate the CRD and NT play important roles during induction of T cell apoptosis, which suggests their potential as therapeutic targets for reversing cancer immune tolerance.
Collapse
|
29
|
Su L, Jiang Y, Xu Y, Li X, Gao W, Xu C, Zeng C, Song J, Weng W, Liang W. Xihuang pill promotes apoptosis of Treg cells in the tumor microenvironment in 4T1 mouse breast cancer by upregulating MEKK1/SEK1/JNK1/AP-1 pathway. Biomed Pharmacother 2018; 102:1111-1119. [PMID: 29710529 DOI: 10.1016/j.biopha.2018.03.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/09/2018] [Accepted: 03/11/2018] [Indexed: 01/14/2023] Open
Abstract
OBJECTIVE To determine the role of the MEKK1/SEK1/JNK1/AP-1 pathway in the action of Xihuang pill (XHP) in reducing regulatory T (Treg) cell numbers in the tumor microenvironment in a 4T1 mouse breast cancer model, and to clarify the anti-tumor mechanism of XHP in breast cancer. METHODS We established a mouse 4T1 breast cancer model. Model mice were administered XHP for 2 weeks, and tumor tissues were then removed, weighed, sliced, and homogenized. Treg cells in the tumor microenvironment were isolated by magnetic cell sorting and analyzed by immunohistochemistry and flow cytometry. Treg cell apoptosis was detected by TdT-mediated dUTP nick end labeling. mRNA expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment were detected by quantitative real-time PCR and their protein expression levels were detected by immunofluorescence staining and western blot. RESULTS Tumor weights were significantly lower in the XHP groups compared with the untreated control group. The overall number of Treg cells in the tumor microenvironment decreased while the number of apoptotic Treg cells increased with increasing doses of XHP. mRNA and protein expression levels of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment increased with increasing doses of XHP. CONCLUSION XHP might promote Treg cell apoptosis in the tumor microenvironment and further inhibit the tumor growth of 4T1 mouse breast cancer. The mechanism of XHP may be related to upregulation of gene and protein expression of MEKK1, SEK1, JNK1, and AP-1 in Treg cells in the tumor microenvironment.
Collapse
Affiliation(s)
- Liang Su
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Yiming Jiang
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Yu Xu
- Medical College of Dalian University, Dalian 116622, China
| | - Xinye Li
- Medical College of Dalian University, Dalian 116622, China
| | - Wenbin Gao
- Department of Medical Oncology, The 3rd Affiliated Hospital of Shenzhen University, Shenzhen 518001, China
| | - Chunwei Xu
- Department of Pathology, Affiliated Hospital of Academy of Military Medical Sciences, Beijing 100071, China
| | - Changqian Zeng
- Medical College of Dalian University, Dalian 116622, China
| | - Jie Song
- Medical College of Dalian University, Dalian 116622, China
| | - Wencai Weng
- Xin Hua Affiliated Hospital of Dalian University, Dalian 116000, China
| | - Wenbo Liang
- Medical College of Dalian University, Dalian 116622, China.
| |
Collapse
|
30
|
Jaiswal S, Srivastava KK. Protein tyrosine kinase A modulates intracellular survival of mycobacteria through Galectin 3. Biochem Biophys Res Commun 2018; 498:884-890. [PMID: 29545176 DOI: 10.1016/j.bbrc.2018.03.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 03/09/2018] [Indexed: 10/17/2022]
Abstract
Mycobacterium tuberculosis (MTB) is a successful pathogen which increases persistence inside the host macrophage by subverting its defence mechanism. Mycobacteria regulate the pathogenesis and intracellular survival by controlling its interaction with host protein(s). Galectin 3 is a member of the β-galactoside binding gene family which is involved in several biological functions. In the present study, we have expressed the mycobacterial protein tyrosine kinase (PtkA) in the cytosol of host macrophages through a eukaryotic promoter vector and found that it down-regulates Galectin 3. Infection by ptkA knocked-out (KO) mycobacterial strain shows increased level of Galectin 3 in the cytosol of macrophages. PtkA regulates Galectin 3 level and stimulates host macrophage through MEK-JNK-cJUN pathway and initiates early apoptosis in H37Ra infected macrophage. The ptkA KO strain showed decreased progression of apoptosis confirming Galectin 3 as anti-apoptotic molecule. The intracellular survival was also found to be impaired in the mice infected with ptkA KO mycobacteria. The hypothesis was also confirmed by looking at the intracellular survival of mycobacteria in Galectin 3 silenced macrophages. The overall findings suggest the significance of Galectin 3 and PtkA interaction in intracellular persistence of mycobacteria.
Collapse
Affiliation(s)
- Swati Jaiswal
- Division of Microbiology and Academy of Scientific and Innovative Research, India
| | | |
Collapse
|
31
|
Vinnai JR, Cumming RC, Thompson GJ, Timoshenko AV. The association between oxidative stress-induced galectins and differentiation of human promyelocytic HL-60 cells. Exp Cell Res 2017; 355:113-123. [DOI: 10.1016/j.yexcr.2017.03.059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 12/14/2022]
|
32
|
Hornung Á, Monostori É, Kovács L. Systemic lupus erythematosus in the light of the regulatory effects of galectin-1 on T-cell function. Lupus 2017; 26:339-347. [PMID: 28100106 DOI: 10.1177/0961203316686846] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Galectin-1 is an endogenous immunoregulatory lectin-type protein. Its most important effects are the inhibition of the differentiation and cytokine production of Th1 and Th17 cells, and the induction of apoptosis of activated T-cells. Galectin-1 has been identified as a key molecule in antitumor immune surveillance, and data are accumulating about the pathogenic role of its deficiency, and the beneficial effects of its administration in various autoimmune disease models. Initial animal and human studies strongly suggest deficiencies in both galectin-1 production and responsiveness in systemic lupus erythematosus (SLE) T-cells. Since lupus features widespread abnormalities in T-cell activation, differentiation and viability, in this review the authors wished to highlight potential points in T-cell signalling processes that may be influenced by galectin-1. These points include GM-1 ganglioside-mediated lipid raft aggregation, early activation signalling steps involving p56Lck, the exchange of the CD3 ζ-ZAP-70 to the FcRγ-Syk pathway, defective mitogen-activated protein kinase pathway activation, impaired regulatory T-cell function, the failure to suppress the activity of interleukin 17 (IL-17) producing T-cells, and decreased suppression of the PI3K-mTOR pathway by phosphatase and tensin homolog (PTEN). These findings place galectin-1 into the group of potential pathogenic molecules in SLE.
Collapse
Affiliation(s)
- Á Hornung
- 1 Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary.,2 Department of Rheumatology and Immunology, University of Szeged, Faculty of Medicine, Albert Szent-Györgyi Health Centre, Szeged, Hungary
| | - É Monostori
- 1 Institute of Genetics, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - L Kovács
- 2 Department of Rheumatology and Immunology, University of Szeged, Faculty of Medicine, Albert Szent-Györgyi Health Centre, Szeged, Hungary
| |
Collapse
|
33
|
|
34
|
Kang SH, Hwang IH, Son E, Cho CK, Choi JS, Park SJ, Jang BC, Lee KB, Lee ZW, Lee JH, Yoo HS, Jang IS. Allergen-Removed Rhus verniciflua Extract Induces Ovarian Cancer Cell Death via JNK Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:1719-1735. [PMID: 27848251 DOI: 10.1142/s0192415x16500968] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Nuclear factor-[Formula: see text]B (NF-[Formula: see text]B)/Rel transcription factors are best known for their central roles in promoting cell survival in cancer. NF-[Formula: see text]B antagonizes tumor necrosis factor (TNF)-[Formula: see text]-induced apoptosis through a process involving attenuation of the c-Jun-N-terminal kinase (JNK). However, the role of JNK activation in apoptosis induced by negative regulation of NF-[Formula: see text]B is not completely understood. We found that allergen-removed Rhus verniciflua Stokes (aRVS) extract-mediated NF-[Formula: see text]B inhibition induces apoptosis in SKOV-3 ovarian cancer cells via the serial activation of caspases and SKOV-3 cells are most specifically suppressed by aRVS. Here, we show that in addition to activating caspases, aRVS extract negatively modulates the TNF-[Formula: see text]-mediated I[Formula: see text]B/NF-[Formula: see text]B pathway to promote JNK activation, which results in apoptosis. When the cytokine TNF-[Formula: see text] binds to the TNF receptor, I[Formula: see text]B dissociates from NF-[Formula: see text]B. As a result, the active NF-[Formula: see text]B translocates to the nucleus. aRVS extract (0.5[Formula: see text]mg/ml) clearly prevented NF-[Formula: see text]B from mobilizing to the nucleus, resulting in the upregulation of JNK phosphorylation. This subsequently increased Bax activation, leading to marked aRVS-induced apoptosis, whereas the JNK inhibitor SP600125 in aRVS extract treated SKOV-3 cells strongly inhibited Bax. Bax subfamily proteins induced apoptosis through caspase-3. Thus, these results indicate that aRVS extract contains components that inhibit NF-[Formula: see text]B signaling to upregulate JNK activation in ovarian cancer cells and support the potential of aRVS as a therapeutic agent for ovarian cancer.
Collapse
Affiliation(s)
- Se-Hui Kang
- * Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea
| | - In-Hu Hwang
- † Department of Physiology, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Eunju Son
- ‡ Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Korea
| | - Chong-Kwan Cho
- § East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Jong-Soon Choi
- * Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea.,‡ Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 305-764, Korea
| | - Soo-Jung Park
- ¶ Department of Sasang Constitutional Medicine, College of Korean Medicine, Woosuk University, Wanju, Jeonbuk, 55338, Republic of Korea
| | - Byeong-Churl Jang
- ∥ Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 704-701, Republic of Korea
| | - Kyung-Bok Lee
- * Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea
| | - Zee-Won Lee
- * Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea
| | - Jong Hoon Lee
- ** Department of Integrative Cancer Center, Woosuk Korean Medicine Hospital, Woosuk University, Jeonju 560-833, Republic of Korea
| | - Hwa-Seung Yoo
- § East-West Cancer Center, Daejeon University, Daejeon 302-120, Republic of Korea
| | - Ik-Soon Jang
- * Division of Bioconvergence, Korea Basic Science Institute, Daejeon 305-333, Republic of Korea
| |
Collapse
|
35
|
Zhang D, Chen Z, Wang DC, Wang X. Regulatory T cells and potential inmmunotherapeutic targets in lung cancer. Cancer Metastasis Rev 2016; 34:277-90. [PMID: 25962964 DOI: 10.1007/s10555-015-9566-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung cancer and metastasis are two of the most lethal diseases globally and seldom have effective therapies. Immunotherapy is considered as one of the powerful alternatives. Regulatory T cells (Tregs) can suppress the activation of the immune system, maintain immune tolerance to self-antigens, and contribute to immunosuppression of antitumor immunity, which is critical for tumor immune evasion in epithelial malignancies, including lung cancer. The present review gives an overview of the biological functions and regulations of Tregs associated with the development of lung cancer and metastasis and explores the potentials of Treg-oriented therapeutic targets. Subsets and features of Tregs mainly include naturally occurring Tregs (nTregs) (CD4(+) nTregs and CD8(+) nTregs) and adaptive/induced Tregs (CD4(+) iTregs and CD8(+) iTregs). Tregs, especially in circulation or regional lymph nodes, play an important role in the progress and metastasis of lung cancer and are considered as therapeutic targets and biomarkers to predict the survival length and recurrence of lung cancer. Increasing understanding of Tregs' functional mechanisms will lead to a number of clinical trials on the discovery and development of Treg-oriented new therapies. Tregs play important roles in lung cancer and metastasis, and the understanding of Tregs becomes more critical for clinical applications and therapies. Thus, Tregs and associated factors can be potential therapeutic targets for lung cancer immunotherapy.
Collapse
Affiliation(s)
- Ding Zhang
- Minhang Hospital, Zhongshan Hospital, Fudan University, Shanghai Institute of Clinical Bioinformatics, Fudan University Center for Clinical Bioinformatics, Shanghai, China
| | | | | | | |
Collapse
|
36
|
Zheng L, Xu C, Guan Z, Su X, Xu Z, Cao J, Teng L. Galectin-1 mediates TGF-β-induced transformation from normal fibroblasts into carcinoma-associated fibroblasts and promotes tumor progression in gastric cancer. Am J Transl Res 2016; 8:1641-1658. [PMID: 27186290 PMCID: PMC4859895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/05/2016] [Indexed: 06/05/2023]
Abstract
Rcinoma-associated fibroblasts (CAFs) are a major constituent of the tumor microenvironment. Cancer cells can induce the transformation from normal fibroblasts (NFs) into CAFs, reciprocally, CAFs promote tumor invasion and proliferation. TGF-β has been the mostly accepted factor to fuel NFs transformation into CAFs. Galectin-1 (Gal1) is highly upregulated in CAFs of multiple human cancers, and overexpression of Gal1 in CAFs promotes tumor progression. The effect of Gal1 on TGF-β-induced CAFs activation has not yet been established in gastric cancer (GC). In this study, we show that Gal1 expression in stroma is positively related to TGF-β in epithelial cells by retrospective analysis of GC patient samples. Meanwhile, conditioned media (CMs) from gastric cancer cells induce expression of both Gal1 and the CAFs marker alpha smooth muscle actin (α-SMA) in NFs via TGF-β secretion. Knockdown of Gal1 prevents TGF-β-induced the conversion of NFs to CAFs. CMs from fibroblasts overexpressing Gal1 inhibits cancer cells apoptosis, promotes migration and invasion in vitro. Thus, Gal1 is significantly involved in the development of tumor-promoting microenvironment by enhancing TGF-β signaling in a positive feedback loop. Targeting Gal1 in tumor stroma should be considered as a potential therapeutic target for GC.
Collapse
Affiliation(s)
- Lingyan Zheng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Cong Xu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Zhonghai Guan
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Xingyun Su
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Zhenzhen Xu
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| | - Jiang Cao
- Clinical Research Center, The Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou, Zhejiang, China
| | - Lisong Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhou, Zhejiang, China
| |
Collapse
|
37
|
Thulasitha WS, Whang I, Umasuthan N, Kang HS, Mothishri MS, Lee S, Qiang W, Noh JK, Lee J. A galectin related protein from Oplegnathus fasciatus: Genomic, molecular, transcriptional features and biological responses against microbial pathogens. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 56:13-24. [PMID: 26615008 DOI: 10.1016/j.dci.2015.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 06/05/2023]
Abstract
Galectins, a family of β-galactoside-binding lectins, are pattern recognition receptors that recognize pathogen-associated molecular patterns and are subsequently involved in the opsonization, phagocytosis, complement activation, and killing of microbes. Here, we report a novel galectin related protein (GRP) identified from rock bream (Oplegnathus fasciatus), designated OfGal like B. The cDNA of OfGal like B is 517 bp with an open reading frame (ORF) of 438 bp, encoding 145 amino acids, with a single carbohydrate recognition domain (CRD). However, only two of the seven critical residues responsible for carbohydrate recognition were identified in the CRD. There was no signal peptide identified in the OfGal like B protein. The genomic structure of OfGal like B, determined using a bacterial artificial chromosome (BAC) genomic library, consists of four exons and three introns. Homology assessment, multiple sequence alignment, and phylogenetic analysis indicated that OfGal like B is an evolutionarily conserved lectin that is closely related to the proto-type galectins. OfGal like B mRNA was constitutively expressed in a wide range of tissues in healthy rock breams. When challenged with bacterial or viral stimulants, OfGal like B was up-regulated in the gills and spleen of rock breams, indicating that it likely plays an important role during bacterial and viral infections. Furthermore, recombinant OfGal like B (rOfGal like B) lacked carbohydrate-binding activity but was able to recognize and agglutinate bacteria, including Streptococcus iniae, Listeria monocytogenes, Vibrio tapetis, Escherichia coli, and Edwardsiella tarda, and a ciliate parasite, Miamiensis avidus. These results collectively suggest that OfGal like B is involved in pathogen recognition and plays a significant role(s) in the innate defense mechanism of rock bream.
Collapse
Affiliation(s)
- William Shanthakumar Thulasitha
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Ilson Whang
- Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Navaneethaiyer Umasuthan
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Hyun-Sil Kang
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - M S Mothishri
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Seongdo Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Wan Qiang
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea
| | - Jae Koo Noh
- Genetics & Breeding Research Center, National Fisheries Research & Development Institute, Geoje, 656-842, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea; Fish Vaccine Development Center, Jeju National University, Jeju Self-Governing Province, 690-756, Republic of Korea.
| |
Collapse
|
38
|
Molecular cloning, expression of a galectin gene in Pacific white shrimp Litopenaeus vannamei and the antibacterial activity of its recombinant protein. Mol Immunol 2015; 67:325-40. [DOI: 10.1016/j.molimm.2015.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 06/12/2015] [Accepted: 06/16/2015] [Indexed: 12/14/2022]
|
39
|
Freire-de-Lima L, Fonseca LM, Oeltmann T, Mendonça-Previato L, Previato JO. The trans-sialidase, the major Trypanosoma cruzi virulence factor: Three decades of studies. Glycobiology 2015. [PMID: 26224786 DOI: 10.1093/glycob/cwv057] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Chagas' disease is a potentially life-threatening disease caused by the protozoan parasite Trypanosoma cruzi. Since the description of Chagas'disease in 1909 extensive research has identified important events in the disease in order to understand the biochemical mechanism that modulates T. cruzi-host cell interactions and the ability of the parasite to ensure its survival in the infected host. Exactly 30 years ago, we presented evidence for the first time of a trans-sialidase activity in T. cruzi (T. cruzi-TS). This enzyme transfers sialic acid from the host glycoconjugates to the terminal β-galactopyranosyl residues of mucin-like molecules on the parasite's cell surface. Thenceforth, many articles have provided convincing data showing that T. cruzi-TS is able to govern relevant mechanisms involved in the parasite's survival in the mammalian host, such as invasion, escape from the phagolysosomal vacuole, differentiation, down-modulation of host immune responses, among others. The aim of this review is to cover the history of the discovery of T. cruzi-TS, as well as some well-documented biological effects encompassed by this parasite's virulence factor, an enzyme with potential attributes to become a drug target against Chagas disease.
Collapse
Affiliation(s)
- L Freire-de-Lima
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21944902, Rio de Janeiro, RJ, Brasil
| | - L M Fonseca
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21944902, Rio de Janeiro, RJ, Brasil
| | - T Oeltmann
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - L Mendonça-Previato
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21944902, Rio de Janeiro, RJ, Brasil
| | - J O Previato
- Laboratório de Glicobiologia, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, 21944902, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
40
|
Kundumani-Sridharan V, Subramani J, Das KC. Thioredoxin Activates MKK4-NFκB Pathway in a Redox-dependent Manner to Control Manganese Superoxide Dismutase Gene Expression in Endothelial Cells. J Biol Chem 2015; 290:17505-19. [PMID: 26028649 DOI: 10.1074/jbc.m115.660365] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Indexed: 11/06/2022] Open
Abstract
The mitogen-activated protein kinase kinase 4 (MKK4) is activated via phosphorylation of Ser-257 and Thr-261 by upstream MAP3Ks and activates JNK and p38 MAPKs in response to cellular stress. We show that thioredoxin (Trx), a cellular redox protein, activates MKK4 via Cys-246 and Cys-266 residues as mutation of these residues renders MKK4 insensitive to phosphorylation by MAP3Ks, TNFα, or Trx. MKK4 is activated in vitro by reduced Trx but not oxidized Trx in the absence of an upstream kinase, suggesting that autophosphorylation of this protein occurs due to reduction of Cys-246 and Cys-266 by Trx. Additionally, mutation of Cys-246 and Cys-266 resulted in loss of kinase activity suggesting that the redox state of Cys-246 and Cys-266 is a critical determinant of MKK4 activation. Trx induces manganese superoxide dismutase (MnSOD) gene transcription by activating MKK4 via redox control of Cys-246 and Cys-266, as mutation of these residues abrogates MKK4 activation and MnSOD expression. We further show that MKK4 activates NFκB for its binding to the MnSOD promoter, which leads to AP-1 dissociation followed by MnSOD transcription. Taken together, our studies show that the redox status of Cys-246 and Cys-266 in MKK4 controls its activities independent of MAP3K, demonstrating integration of the endothelial redox environment to MAPK signaling.
Collapse
Affiliation(s)
- Venkatesh Kundumani-Sridharan
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Jaganathan Subramani
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| | - Kumuda C Das
- From the Department of Anesthesiology and Center for Excellence in Cardiovascular Research, Texas Tech University Health Sciences Center, Lubbock, Texas 79430
| |
Collapse
|
41
|
Lhuillier C, Barjon C, Niki T, Gelin A, Praz F, Morales O, Souquere S, Hirashima M, Wei M, Dellis O, Busson P. Impact of Exogenous Galectin-9 on Human T Cells: CONTRIBUTION OF THE T CELL RECEPTOR COMPLEX TO ANTIGEN-INDEPENDENT ACTIVATION BUT NOT TO APOPTOSIS INDUCTION. J Biol Chem 2015; 290:16797-811. [PMID: 25947381 DOI: 10.1074/jbc.m115.661272] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Indexed: 11/06/2022] Open
Abstract
Galectin-9 (gal-9) is a multifunctional β-galactoside-binding lectin, frequently released in the extracellular medium, where it acts as a pleiotropic immune modulator. Despite its overall immunosuppressive effects, a recent study has reported bimodal action of gal-9 on human resting blood T cells with apoptosis occurring in the majority of them, followed by a wave of activation and expansion of Th1 cells in the surviving population. Our knowledge of the signaling events triggered by exogenous gal-9 in T cells remains limited. One of these events is cytosolic calcium (Ca(2+)) release reported in some murine and human T cells. The aim of this study was to investigate the contribution of Ca(2+) mobilization to apoptotic and nonapoptotic effects of exogenous gal-9 in human T cells. We found that the T cell receptor (TCR)-CD3 complex and the Lck kinase were required for Ca(2+) mobilization but not for apoptosis induction in Jurkat cells. These data were confirmed in human CD4(+) T cells from peripheral blood as follows: a specific Lck chemical inhibitor abrogated Ca(2+) mobilization but not apoptosis induction. Moreover, Lck activity was also required for the production of Th1-type cytokines, i.e. interleukin-2 and interferon-γ, which resulted from gal-9 stimulation in peripheral CD4(+) T cells. These findings indicate that gal-9 acts on T cells by two distinct pathways as follows: one mimicking antigen-specific activation of the TCR with a mandatory contribution of proximal elements of the TCR complex, especially Lck, and another resulting in apoptosis that is independent of this complex.
Collapse
Affiliation(s)
- Claire Lhuillier
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Clément Barjon
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Toshiro Niki
- the Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan, the GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan
| | - Aurore Gelin
- the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France
| | - Françoise Praz
- INSERM, UMR-S 938, Centre de Recherche Saint-Antoine, 75012, Paris, France, the Sorbonne Universités, UPMC Université Paris 06, UMR-S 938, Centre de Recherche Saint-Antoine, 75012, Paris, France
| | - Olivier Morales
- CNRS, UMR 8161 Groupe IRCV, Institut de Biologie de Lille, 1 Rue du Pr. Calmette, 59021 Lille, France
| | - Sylvie Souquere
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, UMR 8122, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France, and
| | - Mitsuomi Hirashima
- the Department of Immunology and Immunopathology, Faculty of Medicine, Kagawa University, Kagawa 761-0793, Japan, the GalPharma Co., Ltd., Takamatsu, Kagawa 761-0301, Japan
| | - Ming Wei
- the Cellvax, Ecole Nationale Vétérinaire d'Alfort, 7 Avenue du Général de Gaulle, 94704 Maisons-Alfort Cedex, France
| | - Olivier Dellis
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, INSERM, UMR-S 757, Bâtiment 440/443, Rue des Adèles, 91405 Orsay, France
| | - Pierre Busson
- From the Université Paris-Sud, 15 Rue Georges Clémenceau, 91400, Orsay, France, the CNRS, UMR 8126, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94805 Villejuif Cedex, France,
| |
Collapse
|
42
|
Isorhapontigenin (ISO) inhibited cell transformation by inducing G0/G1 phase arrest via increasing MKP-1 mRNA Stability. Oncotarget 2015; 5:2664-77. [PMID: 24797581 PMCID: PMC4058035 DOI: 10.18632/oncotarget.1872] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The cancer chemopreventive property of Chinese herb new isolate isorhapontigenin (ISO) and mechanisms underlying its activity have never been explored. Here we demonstrated that ISO treatment with various concentrations for 3 weeks could dramatically inhibit TPA/EGF-induced cell transformation of Cl41 cells in Soft Agar assay, whereas co-incubation of cells with ISO at the same concentrations could elicit G0/G1 cell-cycle arrest without redundant cytotoxic effects on non-transformed cells. Further studies showed that ISO treatment resulted in cyclin D1 downregulation in dose- and time-dependent manner. Our results indicated that ISO regulated cyclin D1 at transcription level via targeting JNK/C-Jun/AP-1 activation. Moreover, we found that ISO-inhibited JNK/C-Jun/AP-1 activation was mediated by both upregulation of MKP-1 expression through increasing its mRNA stability and deactivating MKK7. Most importantly, MKP-1 knockdown could attenuate ISO-mediated suppression of JNK/C-Jun activation and cyclin D1 expression, as well as G0/G1 cell cycle arrest and cell transformation inhibition, while ectopic expression of FLAG-cyclin D1 T286A mutant also reversed ISO-induced G0/G1 cell-cycle arrest and inhibition of cell transformation. Our results demonstrated that ISO is a promising chemopreventive agent via upregulating mkp-1 mRNA stability, which is distinct from its cancer therapeutic effect with downregulation of XIAP and cyclin D1 expression.
Collapse
|
43
|
Yakushina VD, Vasil’eva OA, Novitskii VV, Andreeva NV, Starikova EG, Tashireva LA, Prokhorenko TS, Zima AP, Ryazantseva NV. Effect of Galectin-1 on Apoptosis of CD4+ Lymphocytes Differentiated In Vitro Towards Regulatory T Cells. Bull Exp Biol Med 2014; 156:669-72. [DOI: 10.1007/s10517-014-2422-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Indexed: 12/25/2022]
|
44
|
Rossi AFT, Duarte MC, Poltronieri AB, Valsechi MC, Jorge YC, de-Santi Neto D, Rahal P, Oliani SM, Silva AE. Deregulation of annexin-A1 and galectin-1 expression in precancerous gastric lesions: intestinal metaplasia and gastric ulcer. Mediators Inflamm 2014; 2014:478138. [PMID: 24719523 PMCID: PMC3955591 DOI: 10.1155/2014/478138] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Annexin-A1 (ANXA1/AnxA1) and galectin-1 (LGALS1/Gal-1) are mediators that play an important role in the inflammatory response and are also associated with carcinogenesis. We investigated mRNA and protein expression in precancerous gastric lesions that participate in the progression cascade to gastric cancer, such as intestinal metaplasia (IM) and gastric ulcer (GU). METHODS Quantitative real-time PCR (qPCR) and immunohistochemical techniques were used to analyze the relative quantification levels (RQ) of ANXA1 and LGALS1 mRNA and protein expression, respectively. RESULTS Increased relative expression levels of ANXA1 were found in 100% of cases, both in IM (mean RQ = 6.22 ± 0.06) and in GU (mean RQ = 6.69 ± 0.10). However, the LGALS1 presented basal expression in both groups (IM: mean RQ = 0.35 ± 0.07; GU: mean RQ = 0.69 ± 0.09). Immunohistochemistry revealed significant positive staining for both the AnxA1 and Gal-1 proteins in the epithelial nucleus and cytoplasm as well as in the stroma of the IM and GU groups (P < 0.05) but absence or low immunorectivity in normal mucosa. CONCLUSION Our results bring an important contribution by evidencing that both the AnxA1 and Gal-1 anti-inflammatory proteins are deregulated in precancerous gastric lesions, suggesting their involvement in the early stages of gastric carcinogenesis, possibly due to an inflammatory process in the gastric mucosa.
Collapse
Affiliation(s)
- Ana Flávia Teixeira Rossi
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Márcia Cristina Duarte
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Ayla Blanco Poltronieri
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Marina Curado Valsechi
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Yvana Cristina Jorge
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Dalísio de-Santi Neto
- Legal Medicine Department and Pathology Service, Hospital de Base, Avenida Brigadeiro Faria Lima 5544, 15090-000 São José do Rio Preto, SP, Brazil
| | - Paula Rahal
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Sonia Maria Oliani
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| | - Ana Elizabete Silva
- Department of Biology, São Paulo State University (UNESP), Câmpus São José do Rio Preto, Rua Cristóvão Colombo 2265, 15054-000 São José do Rio Preto, SP, Brazil
| |
Collapse
|
45
|
Galectin-1 has potential prognostic significance and is implicated in clear cell renal cell carcinoma progression through the HIF/mTOR signaling axis. Br J Cancer 2014; 110:1250-9. [PMID: 24496460 PMCID: PMC3950857 DOI: 10.1038/bjc.2013.828] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 11/28/2013] [Accepted: 12/17/2013] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Metastatic clear cell renal cell carcinoma (ccRCC) patients have <9% 5-year survival rate, do not respond well to targeted therapy and eventually develop resistance. A better understanding of molecular pathways of RCC metastasis is the basis for the discovery of novel prognostic markers and targeted therapies. METHODS We investigated the biological impact of galectin-1 (Gal-1) in RCC cell lines by migration and invasion assays. Effect of Gal-1 expression on the mitogen-activated protein kinase pathway was assessed by proteome array. RESULTS Increased expression of Gal-1 increased cell migration while knocking down Gal-1 expression by siRNA resulted in reduced cellular migration (P<0.001) and invasion (P<0.05). Gal-1 overexpression increased phosphorylation of Akt, mTOR and p70 kinase. Upon hypoxia and increased HIF-1α, Gal-1 increased in a dose-dependent manner. We also found miR-22 overexpression resulted in decreased Gal-1 and HIF-1α. Immunohistochemistry analysis showed that high Gal-1 protein expression was associated with larger size tumor (P=0.034), grades III/IV tumors (P<0.001) and shorter disease-free survival (P=0.0013). Using the Cancer Genome Atlas data set, we found that high Gal-1 mRNA expression was associated with shorter overall survival (41 vs 78 months; P<0.01). CONCLUSIONS Our data suggest Gal-1 mediates migration and invasion through the HIF-1α-mTOR signaling axis and is a potential prognostic marker and therapeutic target.
Collapse
|
46
|
Chávez-Galán L, Ramon-Luing LA, Torre-Bouscoulet L, Pérez-Padilla R, Sada-Ovalle I. Pre-exposure of Mycobacterium tuberculosis-infected macrophages to crystalline silica impairs control of bacterial growth by deregulating the balance between apoptosis and necrosis. PLoS One 2013; 8:e80971. [PMID: 24278357 PMCID: PMC3838437 DOI: 10.1371/journal.pone.0080971] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 10/08/2013] [Indexed: 11/18/2022] Open
Abstract
Inhalation of crystalline silica (CS) particles increases the risk of pulmonary tuberculosis; however, the precise mechanism through which CS exposure facilitates Mycobacterium tuberculosis (Mtb) infection is unclear. We speculate that macrophage exposure to CS deregulates the cell death pathways that could explain, at least in part, the association observed between exposure to CS and pulmonary tuberculosis. We therefore established an in vitro model in which macrophages were exposed to CS and then infected with Mtb. Expression of surface markers was analyzed by flow cytometry, JNK1/2, ASK1, caspase 9, P-p38, Bcl-2 and Mcl-1 were analyzed by Western blot, and cytokines by ELISA. Our results show that exposure to CS limits macrophage ability to control Mtb growth. Moreover, this exposure reduced the expression of TLR2, Bcl-2 and Mcl-1, but increased that of JNK1 and ASK1 molecules in the macrophages. Finally, when the pre-exposed macrophages were infected with Mtb, the concentrations of TNFα, IL-1β and caspase-9 expression increased. This pro-inflammatory profile of the macrophage unbalanced the apoptosis/necrosis pathway. Taken together, these data suggest that macrophages exposed to CS are sensitized to cell death by MAPK kinase-dependent signaling pathway. Secretion of TNF-α and IL-1β by Mtb-infected macrophages promotes necrosis, and this deregulation of cell death pathways may favor the release of viable bacilli, thus leading to the progression of tuberculosis.
Collapse
Affiliation(s)
- Leslie Chávez-Galán
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Lucero A. Ramon-Luing
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Luis Torre-Bouscoulet
- Department of Respiratory Physiology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Rogelio Pérez-Padilla
- Department of Respiratory Physiology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
| | - Isabel Sada-Ovalle
- Laboratory of Integrative Immunology, National Institute of Respiratory Diseases Ismael Cosío Villegas, Mexico City, Mexico
- * E-mail:
| |
Collapse
|
47
|
Aan GJ, Hairi HA, Makpol S, Rahman MA, Karsani SA. Differences in protein changes between stress-induced premature senescence and replicative senescence states. Electrophoresis 2013; 34:2209-17. [DOI: 10.1002/elps.201300086] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 01/10/2023]
Affiliation(s)
- Goon Jo Aan
- Department of Biochemistry; Faculty of Medicine; Universiti Kebangsaan Malaysia; Kuala Lumpur; Malaysia
| | - Haryati Ahmad Hairi
- Department of Biochemistry; Faculty of Medicine; Universiti Kebangsaan Malaysia; Kuala Lumpur; Malaysia
| | - Suzana Makpol
- Department of Biochemistry; Faculty of Medicine; Universiti Kebangsaan Malaysia; Kuala Lumpur; Malaysia
| | - Mariati Abdul Rahman
- Department of Clinical Oral Biology; Faculty of Dentistry; Universiti Kebangsaan Malaysia; Kuala Lumpur; Malaysia
| | - Saiful Anuar Karsani
- Institute of Biological Sciences, Faculty of Science and University of Malaya Centre for Proteomics Research (UMCPR); University of Malaya; Kuala Lumpur; Malaysia
| |
Collapse
|
48
|
Ito K, Stannard K, Gabutero E, Clark AM, Neo SY, Onturk S, Blanchard H, Ralph SJ. Galectin-1 as a potent target for cancer therapy: role in the tumor microenvironment. Cancer Metastasis Rev 2013; 31:763-78. [PMID: 22706847 DOI: 10.1007/s10555-012-9388-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The microenvironment of a tumor is a highly complex milieu, primarily characterized by immunosuppression, abnormal angiogenesis, and hypoxic regions. These features promote tumor progression and metastasis, resulting in poor prognosis and greater resistance to existing cancer therapies. Galectin-1 is a β-galactoside binding protein that is abundantly secreted by almost all types of malignant tumor cells. The expression of galectin-1 is regulated by hypoxia-inducible factor-1 (HIF-1) and it plays vital pro-tumorigenic roles within the tumor microenvironment. In particular, galectin-1 suppresses T cell-mediated cytotoxic immune responses and promotes tumor angiogenesis. However, since galectin-1 displays many different activities by binding to a number of diverse N- or O-glycan modified target proteins, it has been difficult to fully understand how galectin-1 supports tumor growth and metastasis. This review explores the importance of galectin-1 and glycan expression patterns in the tumor microenvironment and the potential effects of inhibiting galectin-1 as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Koichi Ito
- School of Medical Science, Griffith Health Institute, Griffith University, Parklands Drive, Southport, Queensland 4222, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lichtenstein RG, Rabinovich GA. Glycobiology of cell death: when glycans and lectins govern cell fate. Cell Death Differ 2013; 20:976-86. [PMID: 23703323 DOI: 10.1038/cdd.2013.50] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/08/2013] [Accepted: 04/16/2013] [Indexed: 02/04/2023] Open
Abstract
Although one typically thinks of carbohydrates as associated with cell growth and viability, glycosylation also has an integral role in many processes leading to cell death. Glycans, either alone or complexed with glycan-binding proteins, can deliver intracellular signals or control extracellular processes that promote initiation, execution and resolution of cell death programs. Herein, we review the role of glycans and glycan-binding proteins as essential components of the cell death machinery during physiologic and pathologic settings.
Collapse
Affiliation(s)
- R G Lichtenstein
- Avram and Stella Goren-Goldstein, Department of Biotechnology Engineering, Faculty of Engineering, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | |
Collapse
|
50
|
Souza-Fonseca-Guimaraes F, Parlato M, de Oliveira RB, Golenbock D, Fitzgerald K, Shalova IN, Biswas SK, Cavaillon JM, Adib-Conquy M. Interferon-γ and granulocyte/monocyte colony-stimulating factor production by natural killer cells involves different signaling pathways and the adaptor stimulator of interferon genes (STING). J Biol Chem 2013; 288:10715-21. [PMID: 23443666 DOI: 10.1074/jbc.m112.435602] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Natural killer (NK) cells are important for innate immunity in particular through the production of IFN-γ and GM-CSF. Both cytokines are important in restoration of immune function of tolerized leukocytes under inflammatory events. The expression of TLRs in NK cells has been widely studied by analyzing the mRNA of these receptors, rarely seeking their protein expression. We previously showed that murine spleen NK cells express TLR9 intracellularly and respond to CpG oligodeoxynucleotide (CpG-ODN) by producing IFN-γ and GM-CSF. However, to get such production the presence of accessory cytokines (such as IL-15 and IL-18) was required, whereas CpG-ODN or accessory cytokines alone did not induce IFN-γ or GM-CSF. We show here that TLR9 overlaps with the Golgi apparatus in NK cells. Furthermore, CpG-ODN stimulation in the presence of accessory cytokines induces the phosphorylation of c-Jun, STAT3, and IκBα. IFN-γ and GM-CSF production requires NF-κB and STAT3 activation as well as Erk-dependent mechanisms for IFN-γ and p38 signaling for GM-CSF. Using knock-out-mice, we show that UNC93b1 and IL-12 (produced by NK cells themselves) are also necessary for IFN-γ and GM-CSF production. IFN-γ production was found to be MyD88- and TLR9-dependent, whereas GM-CSF was TLR9-independent but dependent on STING (stimulator of interferon genes), a cytosolic adaptor recently described for DNA sensing. Our study thereby allows us to gain insight into the mechanisms of synergy between accessory cytokines and CpG-ODN in NK cells. It also identifies a new and alternative signaling pathway for CpG-ODN in murine NK cells.
Collapse
Affiliation(s)
- Fernando Souza-Fonseca-Guimaraes
- Institut Pasteur, Unit of Cytokines and Inflammation, Department Infection et Epidémiologie, 28 rue du Dr Roux, F-75015 Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|