1
|
Alhendi A, Naser SA. In vitro neutralization of IL-6 receptor exacerbates damage to intestinal epithelial cells during Mycobacterium avium paratuberculosis infection. Front Immunol 2024; 15:1412800. [PMID: 39170608 PMCID: PMC11335550 DOI: 10.3389/fimmu.2024.1412800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Like TNFα, IL-6 is upregulated in Crohn's disease (CD) especially in patients associated with Mycobacterium avium paratuberculosis (MAP) infection, and both cytokines have been targeted as a therapeutic option for the treatment of the disease despite the accepted partial response in some patients. Limited response to anti-IL-6 receptor-neutralizing antibodies therapy may be related to the homeostatic dual role of IL-6. In this study, we investigated the effects and the signaling mechanism of IL-6 involved in intestinal epithelial integrity and function during MAP infection using an in vitro model that consists of THP-1, HT-29 and Caco-2 cell lines. Clinically, we determined that plasma samples from MAP-infected CD patients have higher IL-6 levels compared to controls (P-value < 0.001). In CD-like macrophages, MAP infection has significantly upregulated the secretion of IL-6 and the shedding of (IL-6R) from THP-1 macrophages, P-value < 0.05. Intestinal cell lines (Caco-2 and HT-29) were treated with the supernatant of MAP-infected THP-1 macrophages with or without a neutralizing anti-IL-6R antibody. Treating intestinal Caco-2 cells with supernatant of MAP-infected macrophages resulted in significant upregulation of intestinal damage markers including claudin-2 and SERPINE1/PAI-1. Interestingly, blocking IL-6 signaling exacerbated that damage and further increased the levels of the damage markers. In HT-29 cells, MAP infection upregulated MUC2 expression, a protective response that was reversed when IL-6R was neutralized. More importantly, blocking IL-6 signaling during MAP infection rescued damaged Caco-2 cells from MAP-induced apoptosis. The data clearly supports a protective role of IL-6 in intestinal epithelia integrity and function especially in CD patients associated with MAP infection. The findings may explain the ineffective response to anti-IL6 based therapy and strongly support a therapeutic option that restores the physiologic level of IL-6 in patient's plasma. A new treatment strategy based on attenuation of IL-6 expression and secretion in inflammatory diseases should be considered.
Collapse
Affiliation(s)
| | - Saleh A. Naser
- Division of Molecular Microbiology, Burnett School of Biomedical Sciences, College of Medicine. University of Central Florida, Orlando, FL, United States
| |
Collapse
|
2
|
Liang X, Du W, Huang L, Xiang L, Pan W, Yang F, Zheng F, Xie Y, Geng L, Gong S, Xu W. Helicobacter pylori promotes gastric intestinal metaplasia through activation of IRF3-mediated kynurenine pathway. Cell Commun Signal 2023; 21:141. [PMID: 37328804 PMCID: PMC10273570 DOI: 10.1186/s12964-023-01162-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 05/07/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Metabolic reprogramming is a critical event for cell fate and function, making it an attractive target for clinical therapy. The function of metabolic reprogramming in Helicobacter pylori (H. pylori)-infected gastric intestinal metaplasia remained to be identified. METHODS Xanthurenic acid (XA) was measured in gastric cancer cells treated with H. pylori or H. pylori virulence factor, respectively, and qPCR and WB were performed to detect CDX2 and key metabolic enzymes expression. A subcellular fractionation approach, luciferase and ChIP combined with immunofluorescence were applied to reveal the mechanism underlying H. pylori mediated kynurenine pathway in intestinal metaplasia in vivo and in vitro. RESULTS Herein, we, for the first time, demonstrated that H. pylori contributed to gastric intestinal metaplasia characterized by enhanced Caudal-related homeobox transcription factor-2 (CDX2) and mucin2 (MUC2) expression, which was attributed to activation of kynurenine pathway. H. pylori promoted kynurenine aminotransferase II (KAT2)-mediated kynurenine pathway of tryptophan metabolism, leading to XA production, which further induced CDX2 expression in gastric epithelial cells. Mechanically, H. pylori activated cyclic guanylate adenylate synthase (cGAS)-interferon regulatory factor 3 (IRF3) pathway in gastric epithelial cells, leading to enhance IRF3 nuclear translocation and the binding of IRF3 to KAT2 promoter. Inhibition of KAT2 could significantly reverse the effect of H. pylori on CDX2 expression. Also, the rescue phenomenon was observed in gastric epithelial cells treated with H. pylori after IRF3 inhibition in vitro and in vivo. Most importantly, phospho-IRF3 was confirmed to be a clinical positive relationship with CDX2. CONCLUSION These finding suggested H. pylori contributed to gastric intestinal metaplasia through KAT2-mediated kynurenine pathway of tryptophan metabolism via cGAS-IRF3 signaling, targeting the kynurenine pathway could be a promising strategy to prevent gastric intestinal metaplasia caused by H. pylori infection. Video Abstract.
Collapse
Affiliation(s)
- Xinhua Liang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenjun Du
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Ling Huang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Li Xiang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China
| | - Wenxu Pan
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Fangying Yang
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Fengfeng Zheng
- Department of Infectious Diseases, The Affiliated Hospital of Putian University, Putian, 351100, China
| | - Yongwu Xie
- Department of Hematology, Zhuhai Center for Maternal and Child Health Care, Zhuhai, China
| | - Lanlan Geng
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Sitang Gong
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| | - Wanfu Xu
- Department of Gastroenterology, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
- Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Institute of Pediatrics, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
3
|
Sun H, Chen N, Yang X, Xia Y, Wu D. Effects induced by polyethylene microplastics oral exposure on colon mucin release, inflammation, gut microflora composition and metabolism in mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 220:112340. [PMID: 34015635 DOI: 10.1016/j.ecoenv.2021.112340] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/22/2021] [Accepted: 05/11/2021] [Indexed: 06/12/2023]
Abstract
Microplastics are plastic fragments widely distributed in the environment and accumulate in the organisms. However, the research on microplastics effects in mammals is limited. Polyethylene is the main kind of microplastics in the environment. We hypothesized that polyethylene exposure disrupts host intestine metabolism by modifying intestine microflora composition and then lipopolysaccharide (LPS) pathway. Female mice were orally exposed to 0, 0.002 and 0.2 μg/g/d polyethylene microplastics (PE MPs) for 30 days. Colon mucin density was quantized after AB-PAS staining. Mucin 2 (MUC2), inflammatory factors (IL-1β, IL-6, IL-8 and IL-10), short-chain fatty acid receptors (GPR41 and GPR43), LPS receptors (TLR4 and MyD88) and LPS pathway downstream genes (ERK1 and NF-κB) mRNA levels in colon were measured. Feces were collected on the 15th day of exposure for gut microflora analysis. Blood biochemical analysis was performed. Results showed that 0.2 μg/g/d PE MPs exposure significantly decreased colon mucin expression (p < 0.05), decreased IL-1β (p < 0.05) and increased IL-8 and IL-10 levels (p < 0.01 and p < 0.001 respectively). Microflora data showed that in 0.2 μg/g/d PE MPs group the number of Firmicutes decreased and the number of Bacteroides increased (both p < 0.01). Predicted KEGG metabolic pathways by piecrust method indicated that PE MPs enhanced amino acids metabolism in microflora. ERK1 and NF-κB mRNA were significantly lower in 0.2 μg/g/d PE MPs group (both p < 0.001). Blood total protein, albumin and globulin levels significantly increased after 0.2 μg/g/d PE MPs exposure (p < 0.01, p < 0.01 and p < 0.05 respectively). These results indicate that PE MPs exposure induced decreased mucin production, a slight immune response and increased the microflora amino acid metabolism in the mice colon by modifying colon microflora composition. SUMMARY: Polyethylene microplastics exposure decreased colon mucin release and increased amino acid metabolism by modifying colon microflora composition.
Collapse
Affiliation(s)
- Hanqing Sun
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Na Chen
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xiaona Yang
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Di Wu
- State Key Laboratory of Reproductive Medicine, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
4
|
Gundamaraju R, Chong WC. Consequence of distinctive expression of MUC2 in colorectal cancers: How much is actually bad? Biochim Biophys Acta Rev Cancer 2021; 1876:188579. [PMID: 34139275 DOI: 10.1016/j.bbcan.2021.188579] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022]
Abstract
Colorectal cancer (CRC) exhibits complex pathogenesis via compromised intestinal mucosal barrier. It is accepted that goblet cells secrete mucin which line the intestinal mucosal barrier and offer wide range protection and maintain the gut integrity. The principal mucin in the small and large intestine which is Mucin2 (MUC2) is predominantly expressed in the goblet cells which play a pivotal role in intestinal homeostasis. Its disruption is associated with diverse diseases and carcinomas. MUC2 has lately been identified as a principal marker in various mechanisms and secretory cell lineage. While MUC2 expression is regulated by various modulators, alterations in its expression are associated with immunomodulation, differences in tumor immunity and also regulation of microbiota. In the light of current literature, the present review explicates the regulation, functional mechanisms and essential role of MUC2 in colorectal cancer and aids in providing deep understanding of pathogenesis of the disease and also specifies the importance of the MUC2 in gaining more insights about the subtypes of colorectal cancer and how it can succour in approximating the prognosis and survival of the patients.
Collapse
Affiliation(s)
- Rohit Gundamaraju
- ER Stress and Gut Mucosal Immunology Laboratory, School of Health Sciences, University of Tasmania, Launceston, Tasmania 7248, Australia.
| | - Wai Chin Chong
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia; Department of Molecular and Translational Science, School of Medicine, Nursing, and Health Science, Monash University, Clayton, Victoria 3168, Australia
| |
Collapse
|
5
|
Shadbad MA, Hajiasgharzadeh K, Derakhshani A, Silvestris N, Baghbanzadeh A, Racanelli V, Baradaran B. From Melanoma Development to RNA-Modified Dendritic Cell Vaccines: Highlighting the Lessons From the Past. Front Immunol 2021; 12:623639. [PMID: 33692796 PMCID: PMC7937699 DOI: 10.3389/fimmu.2021.623639] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/26/2021] [Indexed: 12/11/2022] Open
Abstract
Although melanoma remains the deadliest skin cancer, the current treatment has not resulted in the desired outcomes. Unlike chemotherapy, immunotherapy has provided more tolerable approaches and revolutionized cancer therapy. Although dendritic cell-based vaccines have minor side effects, the undesirable response rates of traditional approaches have posed questions about their clinical translation. The immunosuppressive tumor microenvironment can be the underlying reason for their low response rates. Immune checkpoints and indoleamine 2,3-dioxygenase have been implicated in the induction of immunosuppressive tumor microenvironment. Growing evidence indicates that the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase/Protein kinase B (PKB) (PI3K/AKT) pathways, as the main oncogenic pathways of melanoma, can upregulate the tumoral immune checkpoints, like programmed death-ligand 1. This study briefly represents the main oncogenic pathways of melanoma and highlights the cross-talk between these oncogenic pathways with indoleamine 2,3-dioxygenase, tumoral immune checkpoints, and myeloid-derived suppressor cells. Moreover, this study sheds light on a novel tumor antigen on melanoma, which has substantial roles in tumoral immune checkpoints expression, indoleamine 2,3-dioxygenase secretion, and stimulating the oncogenic pathways. Finally, this review collects the lessons from the previous unsuccessful trials and integrates their lessons with new approaches in RNA-modified dendritic cell vaccines. Unlike traditional approaches, the advances in single-cell RNA-sequencing techniques and RNA-modified dendritic cell vaccines along with combined therapy of the immune checkpoint inhibitors, indoleamine 2,3-dioxygenase inhibitor, and RNA-modified dendritic cell-based vaccine can overcome these auto-inductive loops and pave the way for developing robust dendritic cell-based vaccines with the most favorable response rate and the least side effects.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/adverse effects
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/therapeutic use
- Cancer Vaccines/adverse effects
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/transplantation
- Humans
- Immune Checkpoint Proteins/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/metabolism
- Melanoma/therapy
- Myeloid-Derived Suppressor Cells/immunology
- Myeloid-Derived Suppressor Cells/metabolism
- RNA, Small Interfering/adverse effects
- RNA, Small Interfering/genetics
- RNA, Small Interfering/immunology
- RNA, Small Interfering/therapeutic use
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/therapy
- Tumor Escape
- Tumor Microenvironment
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/therapeutic use
- mRNA Vaccines
Collapse
Affiliation(s)
- Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| | - Nicola Silvestris
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Kost-Alimova M, Sidhom EH, Satyam A, Chamberlain BT, Dvela-Levitt M, Melanson M, Alper SL, Santos J, Gutierrez J, Subramanian A, Byrne PJ, Grinkevich E, Reyes-Bricio E, Kim C, Clark AR, Watts AJ, Thompson R, Marshall J, Pablo JL, Coraor J, Roignot J, Vernon KA, Keller K, Campbell A, Emani M, Racette M, Bazua-Valenti S, Padovano V, Weins A, McAdoo SP, Tam FW, Ronco L, Wagner F, Tsokos GC, Shaw JL, Greka A. A High-Content Screen for Mucin-1-Reducing Compounds Identifies Fostamatinib as a Candidate for Rapid Repurposing for Acute Lung Injury. Cell Rep Med 2020; 1:100137. [PMID: 33294858 PMCID: PMC7691435 DOI: 10.1016/j.xcrm.2020.100137] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022]
Abstract
Drug repurposing has the advantage of identifying potential treatments on a shortened timescale. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high-content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce mucin-1 (MUC1) protein abundance. Elevated MUC1 levels predict the development of acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) and correlate with poor clinical outcomes. Our screen identifies fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo, fostamatinib reduces MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro, SYK inhibition by the active metabolite R406 promotes MUC1 removal from the cell surface. Our work suggests fostamatinib as a repurposing drug candidate for ALI.
Collapse
Affiliation(s)
| | - Eriene-Heidi Sidhom
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Abhigyan Satyam
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Moran Dvela-Levitt
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Seth L. Alper
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jean Santos
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Juan Gutierrez
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Choah Kim
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Abbe R. Clark
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrew J.B. Watts
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Jamie Marshall
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Juliana Coraor
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Julie Roignot
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Katherine A. Vernon
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Keith Keller
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Alissa Campbell
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | - Silvana Bazua-Valenti
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Astrid Weins
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephen P. McAdoo
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Frederick W.K. Tam
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Luciene Ronco
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - George C. Tsokos
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Anna Greka
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Alimova M, Sidhom EH, Satyam A, Dvela-Levitt M, Melanson M, Chamberlain BT, Alper SL, Santos J, Gutierrez J, Subramanian A, Grinkevich E, Bricio ER, Kim C, Clark A, Watts A, Thompson R, Marshall J, Pablo JL, Coraor J, Roignot J, Vernon KA, Keller K, Campbell A, Emani M, Racette M, Bazua-Valenti S, Padovano V, Weins A, McAdoo SP, Tam FW, Ronco L, Wagner F, Tsokos GC, Shaw JL, Greka A. A High Content Screen for Mucin-1-Reducing Compounds Identifies Fostamatinib as a Candidate for Rapid Repurposing for Acute Lung Injury during the COVID-19 pandemic. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.06.30.180380. [PMID: 32637960 PMCID: PMC7337390 DOI: 10.1101/2020.06.30.180380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/28/2023]
Abstract
Drug repurposing is the only method capable of delivering treatments on the shortened time-scale required for patients afflicted with lung disease arising from SARS-CoV-2 infection. Mucin-1 (MUC1), a membrane-bound molecule expressed on the apical surfaces of most mucosal epithelial cells, is a biochemical marker whose elevated levels predict the development of acute lung injury (ALI) and respiratory distress syndrome (ARDS), and correlate with poor clinical outcomes. In response to the pandemic spread of SARS-CoV-2, we took advantage of a high content screen of 3,713 compounds at different stages of clinical development to identify FDA-approved compounds that reduce MUC1 protein abundance. Our screen identified Fostamatinib (R788), an inhibitor of spleen tyrosine kinase (SYK) approved for the treatment of chronic immune thrombocytopenia, as a repurposing candidate for the treatment of ALI. In vivo , Fostamatinib reduced MUC1 abundance in lung epithelial cells in a mouse model of ALI. In vitro , SYK inhibition by Fostamatinib promoted MUC1 removal from the cell surface. Our work reveals Fostamatinib as a repurposing drug candidate for ALI and provides the rationale for rapidly standing up clinical trials to test Fostamatinib efficacy in patients with COVID-19 lung injury.
Collapse
Affiliation(s)
- Maria Alimova
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Eriene-Heidi Sidhom
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Abhigyan Satyam
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Moran Dvela-Levitt
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Michelle Melanson
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Seth L. Alper
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean Santos
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Juan Gutierrez
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | | | | | - Choah Kim
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Abbe Clark
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Andrew Watts
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rebecca Thompson
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Jamie Marshall
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Juliana Coraor
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julie Roignot
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Katherine A. Vernon
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keith Keller
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Alissa Campbell
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Matthew Racette
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Silvana Bazua-Valenti
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Valeria Padovano
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Astrid Weins
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen P. McAdoo
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Frederick W.K. Tam
- Department of Immunology and Inflammation, Imperial College, Hammersmith Hospital, London, UK
| | - Lucienne Ronco
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Florence Wagner
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - George C. Tsokos
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Jillian L. Shaw
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Anna Greka
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
8
|
Hebeda CB, Sandri S, Benis CM, de Paula-Silva M, Loiola RA, Reutelingsperger C, Perretti M, Farsky SHP. Annexin A1/Formyl Peptide Receptor Pathway Controls Uterine Receptivity to the Blastocyst. Cells 2020; 9:cells9051188. [PMID: 32403233 PMCID: PMC7291299 DOI: 10.3390/cells9051188] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 04/22/2020] [Accepted: 04/28/2020] [Indexed: 12/13/2022] Open
Abstract
Embryo implantation into the uterine wall is a highly modulated, complex process. We previously demonstrated that Annexin A1 (AnxA1), which is a protein secreted by epithelial and inflammatory cells in the uterine microenvironment, controls embryo implantation in vivo. Here, we decipher the effects of recombinant AnxA1 in this phenomenon by using human trophoblast cell (BeWo) spheroids and uterine epithelial cells (Ishikawa; IK). AnxA1-treated IK cells demonstrated greater levels of spheroid adherence and upregulation of the tight junction molecules claudin-1 and zona occludens-1, as well as the glycoprotein mucin-1 (Muc-1). The latter effect of AnxA1 was not mediated through IL-6 secreted from IK cells, a known inducer of Muc-1 expression. Rather, these effects of AnxA1 involved activation of the formyl peptide receptors FPR1 and FPR2, as pharmacological blockade of FPR1 or FPR1/FPR2 abrogated such responses. The downstream actions of AnxA1 were mediated through the ERK1/2 phosphorylation pathway and F-actin polymerization in IK cells, as blockade of ERK1/2 phosphorylation reversed AnxA1-induced Muc-1 and claudin-1 expression. Moreover, FPR2 activation by AnxA1 induced vascular endothelial growth factor (VEGF) secretion by IK cells, and the supernatant of AnxA1-treated IK cells evoked angiogenesis in vitro. In conclusion, these data highlight the role of the AnxA1/FPR1/FPR2 pathway in uterine epithelial control of blastocyst implantation.
Collapse
Affiliation(s)
- Cristina B. Hebeda
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Silvana Sandri
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Cláudia M. Benis
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Marina de Paula-Silva
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Rodrigo A. Loiola
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
| | - Chris Reutelingsperger
- Faculty of Health, Medicine and Life Sciences, Part of Maastricht University Medical Center, Part of Maastricht University, 6211 LK Maastricht, The Netherlands;
| | - Mauro Perretti
- The William Harvey Research Institute, Queen Mary University of London, London EC1M 6BQ, UK;
| | - Sandra H. P. Farsky
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, São Paulo CEP 05508-000, Brazil; (C.B.H.); (S.S.); (C.M.B.); (M.d.P.-S.); (R.A.L.)
- Correspondence: ; Tel.: +55-(11)-3091-2197
| |
Collapse
|
9
|
Lin R, Sun Y, Ye W, Zheng T, Wen J, Deng Y. T-2 toxin inhibits the production of mucin via activating the IRE1/XBP1 pathway. Toxicology 2019; 424:152230. [PMID: 31170431 DOI: 10.1016/j.tox.2019.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/24/2019] [Accepted: 06/03/2019] [Indexed: 01/07/2023]
Abstract
T-2 toxin is a trichothecene mycotoxin that widely contaminates food and has a variety of toxic effects. However, the underlying mechanism of T-2 toxin on intestinal mucin remains unclear. In present study, human intestinal Caco-2 cells and HT-29 cells were treated with 100 ng/mL T-2 toxin at one-quarter of the IC50 for 24 h, which caused the inhibition of MUC2 and adhesion of E. coli O157:H7. We found T-2 toxin induced endoplasmic reticulum stress and activated the IRE1/XBP1 pathway, which may be related to the inhibition of MUC2. Interestingly, T-2 toxin activated IRE1α to inhibit IRE1β, which optimized mucin production. Furthermore, overexpression of IRE1β in the cells apparently alleviated the inhibition of MUC2 caused by T-2 toxin. IRE1α knock-down blocked the down-regulation of IRE1β and MUC2 induced by T-2 toxin. We revealed the critical role of IRE1α in the inhibition of intestinal mucin. This finding was confirmed in BALB/c mice which were exposed to T-2 toxin (0.5 mg/kg bw) for 4 weeks. T-2 toxin activated the IRE1/XBP1 pathway to disrupt intestinal mucin, which lead to the imbalance of gut microbiota and an increased risk of host infection by E. coli O157:H7. T-2 toxin exposure also increased the expressions of pro-inflammatory cytokines IL-1β, IL-6 and TNF-α in mice, which might respond to IRE1α activation. Importantly, IRE1α activation was a therapeutic target for intestinal inflammation caused by T-2 toxin. This study provided a new perspective to understand the intestinal toxicity of T-2 toxin.
Collapse
Affiliation(s)
- Ruqin Lin
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China
| | - Yu Sun
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China
| | - Wenchu Ye
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China
| | - Ting Zheng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China
| | - Jikai Wen
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China.
| | - Yiqun Deng
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, College of Life Sciences, South China Agricultural University, Guangzhou, Guangdong 510642, PR China; Key Laboratory of Zoonosis of the Ministry of Agriculture and Rural Affairs, South China Agricultural University, Guangzhou, Guangdong, 510642, PR China.
| |
Collapse
|
10
|
Thorpe D, Sultani M, Stringer A. Irinotecan induces enterocyte cell death and changes to muc2 and muc4 composition during mucositis in a tumour-bearing DA rat model. Cancer Chemother Pharmacol 2019; 83:893-904. [PMID: 30815720 DOI: 10.1007/s00280-019-03787-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 01/29/2019] [Indexed: 11/25/2022]
Abstract
Irinotecan-induced mucositis is a major oncological problem. Goblet cells secrete mucus, protecting the intestinal mucosa, with secretion altered during mucositis. The enteric nervous system is involved in regulating gut motility and secretion. The aim of this study was to determine whether enteric neural cells and goblet cells are altered following irinotecan treatment. Tumour-bearing Dark Agouti rats were administered a single dose of 175 mg/kg of irinotecan intraperitoneally and 0.01 mg/kg atropine subcutaneously. Experimental and untreated control rats were killed at times 6, 24, 48, 72, 96 and 120 h after treatment. Jejunum and colon samples were formalin fixed. Haematoxylin and eosin staining, Alcian Blue-PAS staining, and immunohistochemistry with S-100 antibody (neural cell marker) were carried out. Statistical analyses were carried out using Kruskal-Wallis test with Dunns post test, Mann Whitney U test, and nonlinear regression. Total goblet cells decreased at 72 h compared with controls in the colon (p < 0.05). The percentage of cavitated goblet cells decreased compared to all other time points at 120 h in the colon. The number of S-100-positive cells in the submucosal plexus decreased in the colon (p = 0.0046) and in the myenteric plexus of the jejunum and colon (p = 0.0058 and p = 0.0022, respectively), on comparing treated with control. Enteric ganglia in the myenteric plexus of the jejunum decreased at 24 h and 96 h. Irinotecan-induced mucositis is associated with increases in mucus secretion and enteric neural cell change. These changes may contribute to the pathophysiology of mucositis through the dysregulation of neural signalling.
Collapse
Affiliation(s)
- Daniel Thorpe
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, 5001, Australia.
| | - Masooma Sultani
- School of Medical Sciences, Adelaide University, Adelaide, 5001, Australia
| | - Andrea Stringer
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, 5001, Australia
| |
Collapse
|
11
|
Li X, Bleumink-Pluym NMC, Luijkx YMCA, Wubbolts RW, van Putten JPM, Strijbis K. MUC1 is a receptor for the Salmonella SiiE adhesin that enables apical invasion into enterocytes. PLoS Pathog 2019; 15:e1007566. [PMID: 30716138 PMCID: PMC6375660 DOI: 10.1371/journal.ppat.1007566] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 02/14/2019] [Accepted: 01/06/2019] [Indexed: 01/08/2023] Open
Abstract
The cellular invasion machinery of the enteric pathogen Salmonella consists of a type III secretion system (T3SS) with injectable virulence factors that induce uptake by macropinocytosis. Salmonella invasion at the apical surface of intestinal epithelial cells is inefficient, presumably because of a glycosylated barrier formed by transmembrane mucins that prevents T3SS contact with host cells. We observed that Salmonella is capable of apical invasion of intestinal epithelial cells that express the transmembrane mucin MUC1. Knockout of MUC1 in HT29-MTX cells or removal of MUC1 sialic acids by neuraminidase treatment reduced Salmonella apical invasion but did not affect lateral invasion that is not hampered by a defensive barrier. A Salmonella deletion strain lacking the SiiE giant adhesin was unable to invade intestinal epithelial cells through MUC1. SiiE-positive Salmonella closely associated with the MUC1 layer at the apical surface, but invaded Salmonella were negative for the adhesin. Our findings uncover that the transmembrane mucin MUC1 is required for Salmonella SiiE-mediated entry of enterocytes via the apical route. The bacterial pathogen Salmonella enterica is one of the most common causes of human foodborne infection affecting millions of people worldwide each year. To establish infection, Salmonella needs to cross the mucus layer and invade intestinal epithelial cells from the apical surface. However, the apical surface of intestinal epithelial cells is covered with a defensive barrier of large glycosylated transmembrane mucins. These large proteins prevent contact between the Salmonella type III secretion needle and the host plasma membrane thereby preventing invasion. We show for the first time that MUC1, one of the intestinal apical transmembrane mucins, facilitates Salmonella invasion. The Salmonella giant adhesin SiiE is the adhesin responsible for engaging MUC1 and the interaction is mediated by glycans on MUC1. We propose that SiiE interacts with MUC1 in a zipper-like manner that involves repetitive domains in both proteins. Adhesin-receptor interactions are essential for bacterial infection of host cells and key factors in determining target tissues and host range of bacteria. The SiiE-MUC1 invasion pathway may explain tropism of different Salmonella strains and provide a novel target for infection intervention and prevention.
Collapse
Affiliation(s)
- Xinyue Li
- Department of Infectious Diseases & Immunology, Utrecht University, Utrecht, The Netherlands
| | | | - Yvette M. C. A. Luijkx
- Department of Infectious Diseases & Immunology, Utrecht University, Utrecht, The Netherlands
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, The Netherlands
| | - Richard W. Wubbolts
- Department of Biochemistry and Cell Biology, Utrecht University, Utrecht, The Netherlands
| | - Jos P. M. van Putten
- Department of Infectious Diseases & Immunology, Utrecht University, Utrecht, The Netherlands
| | - Karin Strijbis
- Department of Infectious Diseases & Immunology, Utrecht University, Utrecht, The Netherlands
- * E-mail:
| |
Collapse
|
12
|
Mucin 2 silencing promotes colon cancer metastasis through interleukin-6 signaling. Sci Rep 2017; 7:5823. [PMID: 28725043 PMCID: PMC5517441 DOI: 10.1038/s41598-017-04952-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/23/2017] [Indexed: 12/19/2022] Open
Abstract
Downregulation of Mucin 2 (MUC2) expression is associated with early carcinogenesis events in colon cancer. MUC2 plays a role in the progression of colon cancer, and reduced MUC2 protein expression correlates with increased interleukin-6 (IL-6) expression. However, the interaction between MUC2 and IL-6 in colorectal cancer metastasis remains unclear. We systematically analyzed MUC2 and IL-6 expression and determined the survival of cancer patients with high or low MUC2 and IL-6 expression using the Oncomine and PrognoScan databases, respectively. This analysis identified downregulation of MUC2 and overexpression of IL-6 in colon cancer but not in normal colon tissue, and this expression pattern was correlated with poor survival of colon cancer patients. We examined the effects of MUC2 on colon cancer metastasis and used vector-mediated application of short hairpin RNA (shRNA) to suppress MUC2 expression. MUC2 suppressed the migration of colon cancer cells in vitro and dramatically diminished liver metastases in vivo. Treatment with IL-6 increased signal transducer and activator of transcription 3 (STAT3) phosphorylation, promoted checkpoint kinase 2 (Chk2) activation, attenuated cAMP response element-binding protein (CREB) phosphorylation, and suppressed E-cadherin protein expression in MUC2-silenced HT-29 cancer cells. Most importantly, MUC2 is a potential prognostic indicator for colon cancer.
Collapse
|
13
|
Huang HL, Wu HY, Chu PC, Lai IL, Huang PH, Kulp SK, Pan SL, Teng CM, Chen CS. Role of integrin-linked kinase in regulating the protein stability of the MUC1-C oncoprotein in pancreatic cancer cells. Oncogenesis 2017; 6:e359. [PMID: 28692035 PMCID: PMC5541713 DOI: 10.1038/oncsis.2017.61] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/08/2017] [Accepted: 06/05/2017] [Indexed: 12/13/2022] Open
Abstract
MUC1-C overexpression has been associated with the progression of pancreatic tumors by promoting the aggressive and metastatic phenotypes. As MUC1 is a STAT3 target gene, STAT3 plays a major role in regulating MUC1-C expression. In this study, we report an alternative mechanism by which integrin-linked kinase (ILK) post-transcriptionally modulates the expression of MUC1-C by maintaining its protein stability in pancreatic cancer cells. We found that ILK acts in concert with STAT3 to facilitate IL-6-mediated upregulation of MUC1-C; ILK depletion was equally effective as STAT3 depletion in abolishing IL-6-induced MUC1-C overexpression without disturbing the phosphorylation or cellular distribution of STAT3. Conversely, ectopic expression of constitutively active ILK increased MUC1-C expression, though this increase was not noted with kinase-dead ILK. This finding suggests the requirement of the kinase activity of ILK in regulating MUC1-C stability, which was confirmed by using the ILK kinase inhibitor T315. Furthermore, our data suggest the involvement of protein kinase C (PKC)δ in mediating the suppressive effect of ILK inhibition on MUC1-C repression. For example, co-immunoprecipitation analysis indicated that ILK depletion-mediated MUC1-C phosphorylation was accompanied by increased phosphorylation of PKCδ at the activation loop Thr-507 and increased binding of PKCδ to MUC1-C. Conversely, ILK overexpression resulted in decreased PKCδ phosphorylation. From a mechanistic perspective, the present finding, together with our recent report that ILK controls the expression of oncogenic KRAS through a regulatory loop, underscores the pivotal role of ILK in promoting pancreatic cancer progression.
Collapse
Affiliation(s)
- H-L Huang
- The PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - H-Y Wu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.,Institute of Biochemical Science, National Taiwan University, Taipei, Taiwan
| | - P-C Chu
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - I-L Lai
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA.,Epigenome Research Center, China Medical University Hospital, Taichung, Taiwan
| | - P-H Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - S K Kulp
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - S-L Pan
- The PhD Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - C-M Teng
- Pharmacological Institute, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - C-S Chen
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH, USA.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
14
|
Influenza virus infection causes neutrophil dysfunction through reduced G-CSF production and an increased risk of secondary bacteria infection in the lung. Virology 2016; 499:23-29. [DOI: 10.1016/j.virol.2016.08.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/25/2016] [Accepted: 08/26/2016] [Indexed: 11/24/2022]
|
15
|
Chalick M, Jacobi O, Pichinuk E, Garbar C, Bensussan A, Meeker A, Ziv R, Zehavi T, Smorodinsky NI, Hilkens J, Hanisch FG, Rubinstein DB, Wreschner DH. MUC1-ARF-A Novel MUC1 Protein That Resides in the Nucleus and Is Expressed by Alternate Reading Frame Translation of MUC1 mRNA. PLoS One 2016; 11:e0165031. [PMID: 27768738 PMCID: PMC5074479 DOI: 10.1371/journal.pone.0165031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 10/05/2016] [Indexed: 01/26/2023] Open
Abstract
Translation of mRNA in alternate reading frames (ARF) is a naturally occurring process heretofore underappreciated as a generator of protein diversity. The MUC1 gene encodes MUC1-TM, a signal-transducing trans-membrane protein highly expressed in human malignancies. Here we show that an AUG codon downstream to the MUC1-TM initiation codon initiates an alternate reading frame thereby generating a novel protein, MUC1-ARF. MUC1-ARF, like its MUC1-TM 'parent’ protein, contains a tandem repeat (VNTR) domain. However, the amino acid sequence of the MUC1-ARF tandem repeat as well as N- and C- sequences flanking it differ entirely from those of MUC1-TM. In vitro protein synthesis assays and extensive immunohistochemical as well as western blot analyses with MUC1-ARF specific monoclonal antibodies confirmed MUC1-ARF expression. Rather than being expressed at the cell membrane like MUC1-TM, immunostaining showed that MUC1-ARF protein localizes mainly in the nucleus: Immunohistochemical analyses of MUC1-expressing tissues demonstrated MUC1-ARF expression in the nuclei of secretory luminal epithelial cells. MUC1-ARF expression varies in different malignancies. While the malignant epithelial cells of pancreatic cancer show limited expression, in breast cancer tissue MUC1-ARF demonstrates strong nuclear expression. Proinflammatory cytokines upregulate expression of MUC1-ARF protein and co-immunoprecipitation analyses demonstrate association of MUC1-ARF with SH3 domain-containing proteins. Mass spectrometry performed on proteins coprecipitating with MUC1-ARF demonstrated Glucose-6-phosphate 1-dehydrogenase (G6PD) and Dynamin 2 (DNM2). These studies not only reveal that the MUC1 gene generates a previously unidentified MUC1-ARF protein, they also show that just like its ‘parent’ MUC1-TM protein, MUC1-ARF is apparently linked to signaling and malignancy, yet a definitive link to these processes and the roles it plays awaits a precise identification of its molecular functions. Comprising at least 524 amino acids, MUC1-ARF is, furthermore, the longest ARF protein heretofore described.
Collapse
Affiliation(s)
- Michael Chalick
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Oded Jacobi
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Edward Pichinuk
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Christian Garbar
- Department of Biopathology, Institut Jean-Godinot, Reims Cedex, France
| | | | - Alan Meeker
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Ravit Ziv
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
| | - Tania Zehavi
- Department of Pathology, Meir Medical Center, Kfar Saba, Israel
| | | | - John Hilkens
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Franz-Georg Hanisch
- Institute of Biochemistry II, Medical Faculty, University of Cologne, Köln, Germany
| | | | - Daniel H. Wreschner
- Department of Cell Research and Immunology, Tel Aviv University, Ramat Aviv, Israel
- * E-mail:
| |
Collapse
|
16
|
Nguyen PM, Putoczki TL, Ernst M. STAT3-Activating Cytokines: A Therapeutic Opportunity for Inflammatory Bowel Disease? J Interferon Cytokine Res 2015; 35:340-50. [PMID: 25760898 PMCID: PMC4426323 DOI: 10.1089/jir.2014.0225] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 01/15/2015] [Indexed: 01/08/2023] Open
Abstract
The gastrointestinal tract is lined by a single layer of epithelial cells that secrete mucus toward the lumen, which collectively separates the immune sentinels in the underlying lamina propria from the intestinal microflora to prevent aberrant immune responses. Inflammatory bowel disease (IBD) describes a group of autoimmune diseases that arise from defects in epithelial barrier function and, as a consequence, aberrant production of inflammatory cytokines. Among these, interleukin (IL)-6, IL-11, and IL-22 are elevated in human IBD patients and corresponding mouse models and, through activation of the JAK/STAT3 pathway, can both propagate and ameliorate disease. In particular, cytokine-mediated activation of STAT3 in the epithelial lining cells affords cellular protection, survival, and proliferation, thereby affording therapeutic opportunities for the prevention and treatment of colitis. In this review, we focus on recent insights gained from therapeutic modulation of the activities of IL-6, IL-11, and IL-22 in models of IBD and advocate a cautionary approach with these cytokines to minimize their tumor-promoting activities on neoplastic epithelium.
Collapse
Affiliation(s)
- Paul M. Nguyen
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Tracy L. Putoczki
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Matthias Ernst
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
17
|
Shan YS, Hsu HP, Lai MD, Yen MC, Fang JH, Weng TY, Chen YL. Suppression of mucin 2 promotes interleukin-6 secretion and tumor growth in an orthotopic immune-competent colon cancer animal model. Oncol Rep 2014; 32:2335-42. [PMID: 25322805 PMCID: PMC4240497 DOI: 10.3892/or.2014.3544] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 09/19/2014] [Indexed: 02/06/2023] Open
Abstract
Mucin 2 (MUC2) is the major secreted mucin of the large intestine and is expressed by adenomas and mucinous carcinomas. Since colon cancer is associated with a proinflammatory microenvironment and dysregulated MUC2 expression, the aim of this study was to characterize the effects of MUC2 gene expression in colon tumor progression using colonic cancer cells. CT26 colon cancer cells were stably transfected with MUC2 siRNA (MUC2 RNAi) or a control construct containing a nonspecific sequence (scrambled RNAi). Expression of MUC2 was significantly decreased in the MUC2 RNAi cell clones. Although MUC2 suppression did not affect the cell growth of colon cancer cells in vitro, MUC2 knockdown promoted tumor growth in an orthotopic colon cancer model in vivo. MUC2 silencing also increased interleukin (IL)-6 secretion by colon cancer cells. IL-6 neutralization attenuated tumor formation by MUC2 RNAi cells; it also increased CD8 T cell infiltration into the peritoneum. Taken together, to the best of our knowledge, this is the first study indicating that the immune response to cancer cells plays an important role in tumor growth regulated by MUC2. Furthermore, given the effects of MUC2 on IL-6 secretion, its targeting may represent a potentially useful strategy to treat colonic carcinomas.
Collapse
Affiliation(s)
- Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Meng-Chi Yen
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Jung-Hua Fang
- Laboratory Animal Center, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Tzu-Yang Weng
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, R.O.C
| | - Yi-Ling Chen
- Department of Senior Citizen Service Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, R.O.C
| |
Collapse
|
18
|
Mechanisms of antitumor and immune-enhancing activities of MUC1/sec, a secreted form of mucin-1. Immunol Res 2014; 57:70-80. [PMID: 24222275 DOI: 10.1007/s12026-013-8451-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mucin 1 (MUC1) is a polymorphic type 1 transmembrane protein found on the apical surface of normal cells lining the lumen of ducts and glands. Mucins are thought to provide mucosal protection from environmental exposures and carcinogens. An altered form of the MUC1 glycoprotein, which is hypoglycosylated, is expressed in several types of human cancers. In our laboratory, we have found that transfection of a murine mammary tumor cell line with a human secreted isoform of MUC1 rendered these DA-3 cells (DA-3/sec) incapable of growing in intact BALB/c mice. In contrast, implantation of DA-3 cells transfected with the human transmembrane isoform of MUC1 (DA-3/TM), resulted in tumor formation and ultimately death of the animals, similar to the DA-3 parental line. Importantly, inoculation of the DA-3/sec cells in immunodeficient nude mice resulted in tumor formation, indicating that the MUC1/sec molecule's antitumor activity is immunologically controlled. In this review, we summarize the studies we have performed to elucidate possible mechanisms for the immune-mediated antitumor effect of MUC1/sec and/or a unique peptide present in this mucin. Understanding these mechanisms may provide new immunotherapeutic approaches that could be used to target different types of cancer.
Collapse
|
19
|
Ernst M, Thiem S, Nguyen PM, Eissmann M, Putoczki TL. Epithelial gp130/Stat3 functions: an intestinal signaling node in health and disease. Semin Immunol 2014; 26:29-37. [PMID: 24434062 DOI: 10.1016/j.smim.2013.12.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 12/23/2013] [Indexed: 12/16/2022]
Abstract
A contiguous intestinal epithelial barrier safeguards against aberrant activation of the immune system and therefore requires molecular mechanisms that ensure effective wound-healing responses. During this processes cytokine-producing myeloid cells serve as rheostats that link the degree of wounding and local inflammation to the epithelial repair response. Likewise, intestinal inflammation is an important factor by which the microenvironment promotes tumorigenesis and the progression of established cancers by facilitating neoplastic cell survival and proliferation. Among the cytokines and chemokines orchestrating this process, those comprising the interleukin (IL) IL6, IL10/IL22 and IL17/IL23 families play a prominent role by virtue of converging on the latent Signal Transducer and Activator of Transcription (Stat)-3. Accordingly, aberrant and persistent Stat3 activation is a frequent observation in cancers of the gastrointestinal tract where it promotes "cancer hallmark capabilities" in the malignant epithelium and suppresses the anti-tumor response of innate and adaptive immune cells. Here, we discuss recent insights arising from situations where persistent activation of the gp130/Stat3 signaling cascades result from excessive abundance of IL6 family cytokines. In particular, we highlight novel and unique roles for IL11 in promoting intestinal wound-healing and, in its corrupted form, enabling and facilitating growth of inflammation-associated and sporadic gastrointestinal tumors.
Collapse
Affiliation(s)
- Matthias Ernst
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia.
| | - Stefan Thiem
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Paul M Nguyen
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Moritz Eissmann
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| | - Tracy L Putoczki
- The Walter and Eliza Hall Institute for Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Australia
| |
Collapse
|
20
|
Chang CYY, Chen Y, Lin WC, Chen CM, Chen CP, Lee SC, Sheu JJC, Tsai FJ. MUC2 polymorphisms are associated with endometriosis development and infertility: a case-control study. BMC MEDICAL GENETICS 2012; 13:15. [PMID: 22417007 PMCID: PMC3338096 DOI: 10.1186/1471-2350-13-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 03/15/2012] [Indexed: 01/10/2023]
Abstract
BACKGROUND Mucins are highly glycosylated proteins protecting and lubricating epithelial surface of respiratory, gastrointestinal and reproductive tracts. Members of the mucin protein family have been suggested to play an important role in development of endometriosis and infertility. This study investigates genetic association of mucin2 (MUC2) with the risk of endometriosis and endometriosis-related infertility. METHODS This case-control study was conducted at China Medical University Hospital, with 195 endometriosis patients and 196 healthy controls enrolled. Genotyping of six SNPs (rs2856111, rs11245936, rs10794288, rs10902088, rs7103978 and rs11245954) within MUC2 gene were performed by using Taqman genotyping assay; individual SNP and haplotype associations with endometriosis and endometriosis-related infertility were assessed by χ² test. RESULTS Endometriosis patients exhibit significantly lower frequency of the rs10794288 C allele, the rs10902088 T allele and the rs7103978 G allele (P = 0.030, 0.013 and 0.040, respectively). In addition, the rs10794288 C allele and the rs10902088 T allele were also less abundant in patients with infertility versus fertile ones (P = 0.015 and 0.024, respectively). Haplotype analysis of the endometriosis associated SNPs in MUC2 also showed significantly association between the most common haplotypes and endometriosis or endometriosis-related infertility. CONCLUSIONS MUC2 polymorphisms, especially rs10794288 and rs10902088, are associated with endometriosis as well as endometriosis-related infertility. Our data present MUC2 as a new candidate involved in development of endometriosis and related infertility in Taiwanese Han women.
Collapse
Affiliation(s)
- Cherry Yin-Yi Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, 2 Yude Road, 40402 Taichung, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Suppression of mucin 2 enhances the proliferation and invasion of LS174T human colorectal cancer cells. Cell Biol Int 2012; 35:1121-9. [PMID: 21605079 DOI: 10.1042/cbi20100876] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Altered expression of MUC2 (mucin 2) is related to tumour development in colorectal cancer. Colorectal mucinous carcinomas are positive for MUC2 expression, whereas MUC2 is down-regulated in non-mucinous adenocarcinomas. In the present study, we down-regulated MUC2 expression by RNAi (RNA interference) and investigated the in vitro and in vivo effects on the proliferation and invasion/migration potential of the LS174T human colorectal cancer cells. The LS174T cell line is a goblet-cell-like colorectal cancer cell line that continuously produces high levels of MUC2. Inhibition of MUC2 expression in vitro by transfection of LS174T cells with the recombinant plasmid pcDNA6.2-GW/EmGFP-miR-MUC2 led to the production of a stably transfected MUC2-RNAi LS174T cell line. The proliferation and invasion/migration of MUC2-RNAi cells in vitro were significantly higher than those in control cells, as assessed by MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide], colony formation and transwell assays. Subcutaneous injection of MUC2-RNAi LS174T cells into nude mice resulted in the development of subcutaneous tumours visible to the naked eye after 1 week. The growth rate of tumours derived from MUC2-RNAi LS174T cells was greater than that of tumours derived from control cells. Ki67 and matrix metalloproteinase-9 proteins were detected by immunohistochemistry in the xenografts. The expression levels of these proteins were higher in the MUC2-RNAi-derived xenografts than in xenografts derived from control cells. Although the role of MUC2 in colorectal tumorigenesis is not fully understood, these results strongly suggest a relationship between the proliferation and invasion of LS174T cells and the expression of MUC2.
Collapse
|
22
|
Bhattacharya S, Mathew G, Ruban E, Epstein DBA, Krusche A, Hillert R, Schubert W, Khan M. Toponome imaging system: in situ protein network mapping in normal and cancerous colon from the same patient reveals more than five-thousand cancer specific protein clusters and their subcellular annotation by using a three symbol code. J Proteome Res 2010; 9:6112-25. [PMID: 20822185 DOI: 10.1021/pr100157p] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In a proof of principle study, we have applied an automated fluorescence toponome imaging system (TIS) to examine whether TIS can find protein network structures, distinguishing cancerous from normal colon tissue present in a surgical sample from the same patient. By using a three symbol code and a power of combinatorial molecular discrimination (PCMD) of 2(21) per subcellular data point in one single tissue section, we demonstrate an in situ protein network structure, visualized as a mosaic of 6813 protein clusters (combinatorial molecular phenotype or CMPs), in the cancerous part of the colon. By contrast, in the histologically normal colon, TIS identifies nearly 5 times the number of protein clusters as compared to the cancerous part (32 009). By subcellular visualization procedures, we found that many cell surface membrane molecules were closely associated with the cell cytoskeleton as unique CMPs in the normal part of the colon, while the same molecules were disassembled in the cancerous part, suggesting the presence of dysfunctional cytoskeleton-membrane complexes. As expected, glandular and stromal cell signatures were found, but interestingly also found were potentially TIS signatures identifying a very restricted subset of cells expressing several putative stem cell markers, all restricted to the cancerous tissue. The detection of these signatures is based on the extreme searching depth, high degree of dimensionality, and subcellular resolution capacity of TIS. These findings provide the technological rationale for the feasibility of a complete colon cancer toponome to be established by massive parallel high throughput/high content TIS mapping.
Collapse
|
23
|
Li YY, Chang JWC, Hsieh LL, Yeh KY. Neutralization of interleukin (IL)-10 released by monocytes/macrophages enhances the up-regulatory effect of monocyte/macrophage-derived IL-6 on expressions of IL-6 and MUC1, and migration in HT-29 colon cancer cells. Cell Immunol 2010; 265:164-71. [PMID: 20851386 DOI: 10.1016/j.cellimm.2010.07.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Revised: 07/21/2010] [Accepted: 07/26/2010] [Indexed: 12/20/2022]
Abstract
The interactions between monocyte-derived IL-6 and IL-10 in colon cancer are unknown. We continued previous work that showed monocyte/macrophage-derived IL-6 induces IL-6 and MUC1 expression in HT-29 cancer cells, and evaluated if IL-10 present in monocyte/macrophage is involved in this IL-6-mediated effect. We treated HT-29 cells with monocyte/macrophage supernatant following neutralization of monocyte/macrophage-released IL-10. Neutralization markedly enhanced monocyte/macrophage-derived IL-6 effects on HT-29 cells including IL-6 and MUC1 production and cell migration. Double blocking of IL-6 and IL-10 in monocyte/macrophage supernatants abolished this enhancement. Western blot analysis of STAT3 phosphorylation showed that this augmented response in HT-29 cells following IL-10 neutralization is probably mediated through enhanced IL-6-induced phosphorylation (Tyr(705)) of STAT3 proteins. Therefore, monocytes/macrophages have the capacity to release the functionally associated cytokines IL-6 and IL-10 whose interactions can account for the pathogenesis and progression of colon cancer.
Collapse
Affiliation(s)
- Ying-Ying Li
- Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung and Chang Gung University, College of Medicine, Taiwan
| | | | | | | |
Collapse
|
24
|
Starsíchová A, Lincová E, Pernicová Z, Kozubík A, Soucek K. TGF-beta1 suppresses IL-6-induced STAT3 activation through regulation of Jak2 expression in prostate epithelial cells. Cell Signal 2010; 22:1734-44. [PMID: 20603212 DOI: 10.1016/j.cellsig.2010.06.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2010] [Revised: 06/23/2010] [Accepted: 06/24/2010] [Indexed: 10/19/2022]
Abstract
Chronic inflammation plays an important role in the initiation and progression of various human diseases including benign prostatic hyperplasia or prostate cancer. Here we show that the proinflammatory cytokine interleukin-6 (IL-6) has prosurvival effects and chronically activates the Jak2/STAT3 signalling pathway in a model of benign prostatic hyperplasia (BPH-1). We demonstrate that the antiinflammatory cytokine transforming growth factor-beta1 (TGF-beta1), which also permanently activates its canonical signalling pathway through SMAD proteins in BPH-1 cells, modifies the effects of IL-6 on cell proliferation. Importantly, TGF-beta1 inhibits IL-6 signal transduction by decreasing the phosphorylation levels of STAT3. This effect is associated with decreased expression of Jak2 at both mRNA and protein levels. Moreover, we showed that TGF-beta1 inhibits IL-6-induced expression of the cancer-associated gene MUC1. These observations demonstrated a novel interaction between TGF-beta1 and IL-6 signalling and suggested another mechanism of how defects in TGF-beta signalling, frequently associated with prostate pathologies, can contribute to the disruption of tissue homeostasis.
Collapse
Affiliation(s)
- Andrea Starsíchová
- Department of Cytokinetics, Institute of Biophysics, Acad Sci Czech Rep, Královopolská 135, CZ-612 65 Brno, Czech Republic
| | | | | | | | | |
Collapse
|
25
|
Huang L, Frampton G, Liang LJ, DeMorrow S. Aberrant DNA methylation profile in cholangiocarcinoma. World J Gastrointest Pathophysiol 2010; 1:23-9. [PMID: 21607139 PMCID: PMC3097943 DOI: 10.4291/wjgp.v1.i2.23] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 05/25/2010] [Accepted: 06/01/2010] [Indexed: 02/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a notoriously lethal epithelial cancer originating from the biliary system. As radical resection offers a poor success rate and limited effective adjuvant modalities exist in its advanced stage, the disease leads to a fairly poor prognosis. As the incidence of CCA is increasing, although the mortality rate remains stable, and few other definite etiologies have yet to be established, renewing our knowledge of its fundamental carcinogenesis is advisable. The latest advances in molecular carcinogenesis have highlighted the roles of epigenetic perturbations and cancer-related inflammation in CCA. This review focuses on the reciprocal effects between aberrant DNA methylation and inflammatory microenvironment in CCA.
Collapse
|
26
|
Choi S, Park YS, Koga T, Treloar A, Kim KC. TNF-α is a key regulator of MUC1, an anti-inflammatory molecule, during airway Pseudomonas aeruginosa infection. Am J Respir Cell Mol Biol 2010; 44:255-60. [PMID: 20448050 DOI: 10.1165/rcmb.2009-0323oc] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Muc1 is a heterodimeric mucin that is expressed on the apical surface of airway epithelial cells as well as hematopoietic cells. Both in vivo and in vitro studies revealed that Muc1 suppresses inflammatory responses induced by Pseudomonas aeruginosa (PA). In this study, we sought to determine, using intact animals (C57BL/6 mice), whether the expression of Muc1 is important during airway PA infection, and how Muc1 levels are controlled during inflammation. Our results showed that: (1) Muc1 levels in the wild-type (WT) mice were initially low, but gradually increased after PA inhalation, reaching a peak on Day 2, remaining elevated until Day 4, and then gradually decreasing to basal levels on Day 7; (2) TNF receptor 1(-/-) mice failed to increase Muc1 levels after PA infection; (3) after PA inhalation, more inflammatory cells were present in the bronchoalveolar lavage fluid from either Muc1(-/-) or TNF receptor(-/-) mice compared with their WT control animals; (4) more apoptotic neutrophils were present in bronchoalveolar lavage fluid from WT mice compared with Muc1(-/-) mice. We conclude that Muc1(-/-) mice are more inflammatory than WT mice during airway PA infection as a result of both an increase in neutrophil influx and a decrease in neutrophil apoptosis. These results suggest that the up-regulation of Muc1 during airway PA infection might be crucial for suppressing excessive and prolonged inflammatory responses, and is induced mainly by TNF-α, the key proinflammatory mediator.
Collapse
Affiliation(s)
- Seongwon Choi
- Immunology and Asthma Program, Lovelace Respiratory Research Institute, Albuquerque, New Mexico, USA
| | | | | | | | | |
Collapse
|