1
|
Peng J, Sun J, Yu Y, Yuan Q, Zhang Y. Integrative multi-omics analysis reveals the role of toll-like receptor signaling in pancreatic cancer. Sci Rep 2025; 15:52. [PMID: 39747201 PMCID: PMC11696379 DOI: 10.1038/s41598-024-84062-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
As one of the most destructive and invasive cancers, pancreatic cancer exhibits complex tumor heterogeneity, which has been a major challenge for clinicians in terms of patient treatment and prognosis. The toll-like receptor (TLR) pathway is closely related to the immune microenvironment within various cancer tissues. To explore the development pattern of pancreatic cancer and find an ideal biomarker, our research has explored the mechanism of the TLR pathway in pancreatic cancer. We collected single-cell expression data from 57,024 cells and transcriptomic data from 945 pancreatic cancer patients, and conducted a series of analyses at both the single-cell and transcriptomic levels. By calculating the TLR pathway score, we clustered pancreatic cancer patients and conducted a series of analyses including metabolic pathways, immune microenvironment, drug sensitivity and so on. In the process of building prognostic models, we screened 33 core genes related to the prognosis of pancreatic cancer, and combined a series of machine learning algorithms to build the prognosis model of pancreatic cancer. We used single cell sequencing to clarify the complex intrinsic relationship between TLR pathway and pancreatic cancer. The strongest TLR signals were observed in macrophages and endothelial cells. With the occurrence of pancreatic cancer, the TLR signal of various cell types gradually increased, but with the increase of the malignant degree of ductal epithelial cells, the TLR signal gradually weakened. Cluster analysis showed that patients with the most active TLR pathway had severe dysregulation of immune microenvironment and the worst prognosis. Finally, we combined a series of machine learning algorithms to build a pancreatic cancer prognosis model that includes four genes (NT5E, TGFBI, ANLN, and FAM83A). The model showed strong performance in predicting the survival state of pancreatic cancer samples. We explored the important role of TLR pathway in pancreatic cancer and established and validated a new prognosis model for pancreatic cancer based on TLR-related genes.
Collapse
Affiliation(s)
- Jie Peng
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Jiaao Sun
- First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Youfeng Yu
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China
| | - Qihang Yuan
- First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Yong Zhang
- Ningde Clinical Medical College of Fujian Medical University, Fujian, China.
- Ningde Municipal Hospital of Ningde Normal University, Fujian, China.
| |
Collapse
|
2
|
Liu DS, Puik JR, Venø MT, Mato Prado M, Rees E, Patel BY, Merali N, Galloway D, Chan G, Phillips N, Wadsworth C, Vlavianos P, Potts J, Sivakumar S, Davidson BR, Besselink MG, Swijnenburg RJ, Jiao LR, Kazemier G, Giovannetti E, Krell J, Frampton AE. MicroRNAs as Bile-based biomarkers in pancreaticobiliary cancers (MIRABILE): a cohort study. Int J Surg 2024; 110:6518-6527. [PMID: 39041944 PMCID: PMC11486953 DOI: 10.1097/js9.0000000000001888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/16/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Biliary obstruction can be due to both malignant and benign pancreaticobiliary disease. Currently, there are no biomarkers that can accurately help make this distinction. MicroRNAs (miRNAs) are stable molecules in tissue and biofluids that are commonly deregulated in cancer. The MIRABILE study aimed to identify miRNAs in bile that can differentiate malignant from benign pancreaticobiliary disease. MATERIALS AND METHODS There were 111 patients recruited prospectively at endoscopic retrograde cholangiopancreatography (ERCP) or percutaneous transhepatic cholangiography (PTC) for obstructive jaundice, and bile was aspirated for cell-free RNA (cfRNA) extraction and analysis. In a discovery cohort of 78 patients (27 with pancreatic ductal adenocarcinoma (PDAC), 14 cholangiocarcinoma (CCA), 37 benign disease), cfRNA was subjected to small-RNA sequencing. LASSO regression was used to define bile miRNA signatures, and NormFinder to identify endogenous controls. In a second cohort of 87 patients (34 PDAC, 14 CCA, 39 benign disease), RT-qPCR was used for validation. RESULTS LASSO regression identified 14 differentially-expressed bile miRNAs of which 6 were selected for validation. When comparing malignant and benign pancreaticobiliary disease, bile miR-340 and miR-182 were validated and significantly differentially expressed ( P <0.05 and P <0.001, respectively). This generated an AUC of 0.79 (95% CI: 0.70-0.88, sensitivity 65%; specificity 82%) in predicting malignant disease. CONCLUSION Bile collected during biliary drainage contains miRNAs able to differentiate benign from malignant pancreaticobiliary diseases in patients with obstructive jaundice. These bile miRNAs have the potential to increase diagnostic accuracy.
Collapse
Affiliation(s)
- Daniel S.K. Liu
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Jisce R. Puik
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Morten T. Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University, Aarhus C
- Omiics ApS, Aarhus N, Aarhus, Denmark
| | - Mireia Mato Prado
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Eleanor Rees
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Bhavik Y. Patel
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, Section of Oncology, The Leggett Building, University of Surrey
- HPB Surgical Unit, Royal Surrey NHS Foundation Trust, Guildford, Surrey
| | - Nabeel Merali
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, Section of Oncology, The Leggett Building, University of Surrey
- HPB Surgical Unit, Royal Surrey NHS Foundation Trust, Guildford, Surrey
| | - Daniel Galloway
- Department of Gastroenterology, Chelsea and Westminster Hospital, Chelsea and Westminster Hospital NHS Foundation Trust, London
- Department of Gastroenterology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London, W12 0HS
| | - Grace Chan
- Department of Gastroenterology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London, W12 0HS
| | - Natalie Phillips
- Department of Gastroenterology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London, W12 0HS
| | - Christopher Wadsworth
- Department of Gastroenterology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London, W12 0HS
| | - Panagiotis Vlavianos
- Department of Gastroenterology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, Du Cane Road, London, W12 0HS
| | - Jonathan Potts
- Royal Free Sheila Sherlock Liver Centre, Royal Free Hospital and UCL Institute of Liver and Digestive Health, London
| | - Shivan Sivakumar
- Department of Oncology, Institute of Immunology and Immunotherapy, Birmingham Medical School, University of Birmingham, Birmingham
| | - Brian R. Davidson
- Department of HPB and Liver Transplant Surgery, Royal Free Hospital
- Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Marc G. Besselink
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef, Amsterdam, The Netherlands
| | - Rutger-Jan Swijnenburg
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Department of Surgery, Amsterdam UMC location University of Amsterdam, Meibergdreef, Amsterdam, The Netherlands
| | - Long R. Jiao
- Department of Surgery and Oncology, The Royal Marsden Hospital, London, UK
| | - Geert Kazemier
- Department of Surgery, Amsterdam UMC Location Vrije Universiteit Amsterdam
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Jonathan Krell
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Adam E. Frampton
- Department of Clinical and Experimental Medicine, Faculty of Health and Medical Sciences, Section of Oncology, The Leggett Building, University of Surrey
- HPB Surgical Unit, Royal Surrey NHS Foundation Trust, Guildford, Surrey
| |
Collapse
|
3
|
Anılır E, Özen F, Yıldırım İH. Effects of toll like receptor 4 (TLR4) and toll like receptor 2 (TLR 2) gene polymorphisms on clinical outcomes in acute non-biliary pancreatitis patients. Front Immunol 2024; 15:1427187. [PMID: 39221252 PMCID: PMC11361925 DOI: 10.3389/fimmu.2024.1427187] [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: 05/03/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction In this retrospective study, it was aimed to evaluate effects of Toll Like Receptor 4 (TLR4) and Toll Like Receptor 2 (TLR 2) gene polymorphisms on clinical outcomes in acute non-biliary pancreatitis patients. Methods A total of 108 acute non-biliary pancreatitis patients (ANBP) were retrospectively subjected to the study. Gender, age, number of attacks, hospitalization duration, amylase, lipase, aspartate aminotransferase (AST), alanine aminotransferase (ALT), leukocyte, C-reactive protein (CRP), total bilirubin, direct bilirubin, Atlanta score, ultrasonography (USG), Computer Tomography (CT) and patient outcome differences between TLR 4 Rs4986790, TLR 4 Rs4986791 and TLR 2 groups were evaluated. Results According to TLR 4 Rs4986790 groups, females were significantly common in AA sequence (AA) group with statistically significant difference (p<0.05). Leukocyte mean of AG sequence (AG) group was significantly higher than of AA group (p<0.05). All parameter differences between TLR 4 Rs4986791 and TLR 2 groups were statistically insignificant (p>0.05). there was a statistically significant correlation between TLR 4 Rs4986790 and gender (r=0.265; p<0.01), Leukocyte (r=0.200; p<0.05) and Pseudocyst (r=0.203; p<0.05). TLR 4 Rs4986790 gene polymorphism had significant effect on leukocyte level in acute non-biliary pancreatitis patients (OR: -0.1.900; p<0.05). Predictive value of leukocyte for TLR 4 Rs4986790 was statistically significant (Area Under Curve: 0.624; p<0.05). For 7.65 leukocyte cut off value, sensitivity for AA gene polymorphism was 84.2% and specificity was 40.5. Conclusion Although the clinical and outcome parameters of ANBP patients in terms of TLR 4 Rs4986791 and TLR 2 do not show significant differences, research findings point to the diagnostic value of patients' leukocyte parameters in determining TLR-4 Rs4986790 ploimorphism groups.
Collapse
Affiliation(s)
- Ender Anılır
- General Surgery, Biruni University Hospital Organ Transplantation and Hepatobiliary Surgery Center, İstanbul, Türkiye
| | - Filiz Özen
- Medical Genetics, Istanbul Medeniyet University Göztepe Training and Research Hospital Genetic Department, Istanbul, Türkiye
| | - İbrahim Halil Yıldırım
- Department of Genetics, Faculty of Veterinary Medicine, Dicle University, Diyarbakır, Türkiye
| |
Collapse
|
4
|
Banesh S, Patil N, Chethireddy VR, Bhukmaria A, Saudagar P. Design and evaluation of a multiepitope vaccine for pancreatic cancer using immune-dominant epitopes derived from the signature proteome in expression datasets. Med Oncol 2024; 41:90. [PMID: 38522058 DOI: 10.1007/s12032-024-02334-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/14/2024] [Indexed: 03/25/2024]
Abstract
Pancreatic cancer is a highly aggressive and often lethal malignancy with limited treatment options. Its late-stage diagnosis and resistance to conventional therapies make it a significant challenge in oncology. Immunotherapy, particularly cancer vaccines, has emerged as a promising avenue for treating pancreatic cancer. Multi-epitope vaccines, designed to target multiple epitopes derived from various antigens associated with pancreatic cancer, have gained attention as potential candidates for improving therapeutic outcomes. In this study, we have explored transcriptomics and protein expression databases to identify potential upregulated proteins in pancreatic cancer cells. After examining a total of 21,054 proteins from various databases, it was discovered that 143 proteins expressed differently in malignant and healthy cells. The CTL, HTL and BCE epitopes were predicted for the shortlisted proteins. 51,840 vaccine constructs were created by concatenating CTL, HTL, and B-cell epitopes in the respective sequences. The best 86 structures were selected from a set of 51,840 designs after they were analyzed for vaxijenicity, allergenicity, toxicity, and antigenicity scores. In further simulation of the immune response using constructs, it was found that 41417, 37961, and 40841 constructs could produce a strong immune response when injected. Further, it was found that construct 37961 showed stronger interaction and stability with TLR-9 as determined from the large-scale molecular dynamics simulations. Moreover, the 37961 construct has shown interactions with TLR-9 suggests its potential in inducing immune response. In addition, construct 37961 has shown 100% predicted solubility in the E. coli expression system. Overall, the study indicates the designed construct 37961 has the potential to induce an anti-tumor immune response and long-standing protection pending further studies.
Collapse
Affiliation(s)
- Sooram Banesh
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Nupoor Patil
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Vihadhar Reddy Chethireddy
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Arnav Bhukmaria
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India
| | - Prakash Saudagar
- Department of Biotechnology, National Institute of Technology-Warangal, Warangal, Telangana, 506004, India.
| |
Collapse
|
5
|
Ajay AK, Gasser M, Hsiao LL, Böldicke T, Waaga-Gasser AM. TLR2 and TLR9 Blockade Using Specific Intrabodies Inhibits Inflammation-Mediated Pancreatic Cancer Cell Growth. Antibodies (Basel) 2024; 13:11. [PMID: 38390872 PMCID: PMC10885114 DOI: 10.3390/antib13010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/22/2023] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) remains a deadly cancer worldwide with a need for new therapeutic approaches. A dysregulation in the equilibrium between pro- and anti-inflammatory responses with a predominant immunosuppressive inflammatory reaction in advanced stage tumors seem to contribute to tumor growth and metastasis. The current therapies do not include strategies against pro-tumorigenic inflammation in cancer patients. We have shown that the upregulated cell surface expression of Toll-like Receptor (TLR) 2 and of TLR9 inside PDAC cells maintain chronic inflammatory responses, support chemotherapeutic resistance, and mediate tumor progression in human pancreatic cancer. We further demonstrated intracellular TLR2 and TLR9 targeting using specific intrabodies, which resulted in downregulated inflammatory signaling. In this study, we tested, for the first time, an intrabody-mediated TLR blockade in human TLR2- and TLR9-expressing pancreatic cancer cells for its effects on inflammatory signaling-mediated tumor growth. Newly designed anti-TLR2- and anti-TLR9-specific intrabodies inhibited PDAC growth. Co-expression analysis of the intrabodies and corresponding human TLRs showed efficient retention and accumulation of both intrabodies within the endoplasmic reticulum (ER), while co-immunoprecipitation studies indicated both intrabodies interacting with their cognate TLR antigen within the pancreatic cancer cells. Cancer cells with attenuated proliferation expressing accumulated TLR2 and TRL9 intrabodies demonstrated reduced STAT3 phosphorylation signaling, while apoptotic markers Caspases 3 and 8 were upregulated. To conclude, our results demonstrate the TLR2 and TLR9-specific intrabody-mediated signaling pathway inhibition of autoregulatory inflammation inside cancer cells and their proliferation, resulting in the suppression of pancreatic tumor cell growth. These findings underscore the potential of specific intrabody-mediated TLR inhibition in the ER relevant for tumor growth inhibition and open up a new therapeutic intervention strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Amrendra K Ajay
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Martin Gasser
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Li-Li Hsiao
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Thomas Böldicke
- Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Ana Maria Waaga-Gasser
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
6
|
Baima G, Ribaldone DG, Romano F, Aimetti M, Romandini M. The Gum-Gut Axis: Periodontitis and the Risk of Gastrointestinal Cancers. Cancers (Basel) 2023; 15:4594. [PMID: 37760563 PMCID: PMC10526746 DOI: 10.3390/cancers15184594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/01/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Periodontitis has been linked to an increased risk of various chronic non-communicable diseases, including gastrointestinal cancers. Indeed, dysbiosis of the oral microbiome and immune-inflammatory pathways related to periodontitis may impact the pathophysiology of the gastrointestinal tract and its accessory organs through the so-called "gum-gut axis". In addition to the hematogenous spread of periodontal pathogens and inflammatory cytokines, recent research suggests that oral pathobionts may translocate to the gastrointestinal tract through saliva, possibly impacting neoplastic processes in the gastrointestinal, liver, and pancreatic systems. The exact mechanisms by which oral pathogens contribute to the development of digestive tract cancers are not fully understood but may involve dysbiosis of the gut microbiome, chronic inflammation, and immune modulation/evasion, mainly through the interaction with T-helper and monocytic cells. Specifically, keystone periodontal pathogens, including Porphyromonas gingivalis and Fusobacterium nucleatum, are known to interact with the molecular hallmarks of gastrointestinal cancers, inducing genomic mutations, and promote a permissive immune microenvironment by impairing anti-tumor checkpoints. The evidence gathered here suggests a possible role of periodontitis and oral dysbiosis in the carcinogenesis of the enteral tract. The "gum-gut axis" may therefore represent a promising target for the development of strategies for the prevention and treatment of gastrointestinal cancers.
Collapse
Affiliation(s)
- Giacomo Baima
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | | | - Federica Romano
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Aimetti
- Department of Surgical Sciences, University of Turin, 10125 Torino, Italy; (G.B.); (F.R.); (M.A.)
| | - Mario Romandini
- Department of Periodontology, Faculty of Dentistry, University of Oslo, 0313 Oslo, Norway
| |
Collapse
|
7
|
Chai Y, Huang Z, Shen X, Lin T, Zhang Y, Feng X, Mao Q, Liang Y. Microbiota Regulates Pancreatic Cancer Carcinogenesis through Altered Immune Response. Microorganisms 2023; 11:1240. [PMID: 37317214 PMCID: PMC10221276 DOI: 10.3390/microorganisms11051240] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/02/2023] [Accepted: 05/04/2023] [Indexed: 06/16/2023] Open
Abstract
The microbiota is present in many parts of the human body and plays essential roles. The most typical case is the occurrence and development of cancer. Pancreatic cancer (PC), one of the most aggressive and lethal types of cancer, has recently attracted the attention of researchers. Recent research has revealed that the microbiota regulates PC carcinogenesis via an altered immune response. Specifically, the microbiota, in several sites, including the oral cavity, gastrointestinal tract, and pancreatic tissue, along with the numerous small molecules and metabolites it produces, influences cancer progression and treatment by activating oncogenic signaling, enhancing oncogenic metabolic pathways, altering cancer cell proliferation, and triggering chronic inflammation that suppresses tumor immunity. Diagnostics and treatments based on or in combination with the microbiota offer novel insights to improve efficiency compared with existing therapies.
Collapse
Affiliation(s)
- Yihan Chai
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Zhengze Huang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Xuqiu Shen
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Tianyu Lin
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Yiyin Zhang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Xu Feng
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
| | - Qijiang Mao
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
- Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Hangzhou 310016, China
- Zhejiang Province Medical Research Center of Minimally Invasive Diagnosis and Treatment of Abdominal Diseases, Hangzhou 310028, China
| | - Yuelong Liang
- Department of General Surgery, Zhejiang University School of Medicine, Sir Run Run Shaw Hospital, Hangzhou 310016, China
- Zhejiang Province Medical Research Center of Minimally Invasive Diagnosis and Treatment of Abdominal Diseases, Hangzhou 310028, China
| |
Collapse
|
8
|
Nurmi AM, Hagström J, Mustonen H, Seppänen H, Haglund C. The expression and prognostic value of toll-like receptors (TLRs) in pancreatic cancer patients treated with neoadjuvant therapy. PLoS One 2022; 17:e0267792. [PMID: 35536778 PMCID: PMC9089880 DOI: 10.1371/journal.pone.0267792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 04/15/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Toll-like receptors (TLRs) play a pivotal role in the immune system and carcinogenesis. There is no research on TLR expression and association with survival among preoperatively treated pancreatic cancer patients. We studied the expression intensity and prognostic value of TLRs in pancreatic cancer patients treated with neoadjuvant therapy (NAT) and compared the results to patients undergoing upfront surgery (US). METHOD Between 2000 and 2015, 71 borderline resectable patients were treated with NAT and surgery and 145 resectable patients underwent upfront surgery at Helsinki University Hospital, Finland. We immunostained TLRs 1-5, 7, and 9 on sections of tissue-microarray. We classified TLR expression as 0 (negative), 1 (mild), 2 (moderate), or 3 (strong) and divided into high (2-3) and low (0-1) expression for statistical purposes. RESULTS Among TLRs 1, 3, and 9 (TLR1 81% vs 70%, p = 0.008; TLR3 92% vs 68%, p = 0.001; TLR9 cytoplasmic 83% vs 42%, p<0.001; TLR9 membranous 53% vs 25%, p = 0.002) NAT patients exhibited a higher immunopositivity score more frequently than patients undergoing upfront surgery. Among NAT patients, a high expression of TLR1 [Hazards ratio (HR) 0.48, p<0.05] associated with a longer postoperative survival, whereas among US patients, high expression of TLR5 (HR 0.64, p<0.05), TLR7 (HR 0.59, p<0.01, and both TLR7 and TLR9 (HR 0.5, p<0.01) predicted a favorable postoperative outcome in separate analysis adjusted for background variables. CONCLUSIONS We found higher immunopositive intensities among TLRs 1, 3, and 9 in NAT patients. A high TLR1 expression associated with a longer survival among NAT patients, however, among US patients, high expression intensity of TLR5 and TLR7 predicted a favorable postoperative outcome in the adjusted analysis.
Collapse
Affiliation(s)
- Anna Maria Nurmi
- Department of Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Jaana Hagström
- Department of Pathology, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Department of Oral Pathology and Radiology, University of Turku, Turku, Finland
| | - Harri Mustonen
- Department of Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
| | - Hanna Seppänen
- Department of Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, Helsinki University Hospital, University of Helsinki, Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Topcu KSB, Korucu EN, Menevse E, Kocak N, Duran T. Investigation of the effects of the toll-like receptor 4 pathway on immune checkpoint vista in pancreatic cancer. Invest New Drugs 2022; 40:519-528. [PMID: 35113284 DOI: 10.1007/s10637-021-01209-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/28/2021] [Indexed: 12/31/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most common malignant tumors of the pancreas. Preclinical studies show that it evades the immune system with immune checkpoints and promotes tumor development. V-domain Ig suppressor of T cell activation (VISTA) is a new immune-check point from the B7 family and is highly expressed in cancer cells. Overexpression of toll like receptor 4 (TLR4) in pancreatic adenocarcinoma is associated with induced tumorigenesis, tumor growth, resistancy to chemotherapy. Naloxone is an opioid and inhibits TLR4-ligand association. In this study, we investigated the relation of TLR4 and downstream pathways with immune-check point VISTA in pancreatic cancer proliferation. We initially collected pancreatic cancer-related datasets using the GEPIA2 and UALCAN databases. Based on this data obtained the effect of various concentrations and incubation times of naloxone were used on PANC-1 cells proliferation. A combination of naloxone and VISTA-siRNA were applied, and the effect of both naloxone and combined treatment on TLR4, Interleukin 1 receptor associated kinase 4 (IRAK4) and VISTA gene expression were analyzed in pancreatic cancer cells. As a result of analysis with Quantitative Real-Time Polymerase Chain Reaction (qRT-PCR), gene expression levels of TLR4, IRAK4 and VISTA were significantly suppressed and cell proliferation was significantly reduced. We found that administration of naloxone and VISTA-siRNA in combination with PDAC cells suppressed signaling. Therefore, we considered that the relationship between VISTA and TLR4 signaling pathways and the other possible associated signal molecules may be an important marker in determining the response of immune checkpoint inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Kubra Sena Bas Topcu
- Department of Molecular Biology and Genetics, Faculty of Science, Bartın University, Bartın, Turkey
| | - Emine Nedime Korucu
- Department of Molecular Biology and Genetics, Faculty of Science, Necmettin Erbakan University, Konya, Turkey.
| | - Esma Menevse
- Department of Medical Biochemistry, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Nadir Kocak
- Department of Medical Genetics, Faculty of Medicine, Selcuk University, Konya, Turkey
| | - Tugce Duran
- Department of Medical Genetics, Faculty of Medicine, KTO Karatay University, Konya, Turkey
| |
Collapse
|
10
|
Orlacchio A, Mazzone P. The Role of Toll-like Receptors (TLRs) Mediated Inflammation in Pancreatic Cancer Pathophysiology. Int J Mol Sci 2021; 22:12743. [PMID: 34884547 PMCID: PMC8657588 DOI: 10.3390/ijms222312743] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal forms of cancer, characterized by its aggressiveness and metastatic potential. Despite significant improvements in PC treatment and management, the complexity of the molecular pathways underlying its development has severely limited the available therapeutic opportunities. Toll-like receptors (TLRs) play a pivotal role in inflammation and immune response, as they are involved in pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Activation of TLRs initiates a signaling cascade, which in turn, leads to the transcription of several genes involved in inflammation and anti-microbial defense. TLRs are also deregulated in several cancers and can be used as prognostic markers and potential targets for cancer-targeted therapy. In this review we discuss the current knowledge about the role of TLRs in PC progression, focusing on the available TLRs-targeting compounds and their possible use in PC therapy.
Collapse
Affiliation(s)
- Arturo Orlacchio
- NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Pellegrino Mazzone
- Biogem Scarl, Istituto di Ricerche Genetiche Gaetano Salvatore, 83031 Ariano Irpino, Italy
| |
Collapse
|
11
|
Lundy J, Gearing LJ, Gao H, West AC, McLeod L, Deswaerte V, Yu L, Porazinski S, Pajic M, Hertzog PJ, Croagh D, Jenkins BJ. TLR2 activation promotes tumour growth and associates with patient survival and chemotherapy response in pancreatic ductal adenocarcinoma. Oncogene 2021; 40:6007-6022. [PMID: 34400766 DOI: 10.1038/s41388-021-01992-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has an extremely poor prognosis, and is plagued by a paucity of targeted treatment options and tumour resistance to chemotherapeutics. The causal link between chronic inflammation and PDAC suggests that molecular regulators of the immune system promote disease pathogenesis and/or therapeutic resistance, yet their identity is unclear. Here, we couple endoscopic ultrasound-guided fine-needle aspiration, which captures tumour biopsies from all stages, with whole transcriptome profiling of PDAC patient primary tumours to reveal enrichment of the innate immune Toll-like receptor 2 (TLR2) molecular pathway. Augmented TLR2 expression associated with a 4-gene "TLR2 activation" signature, and was prognostic for survival and predictive for gemcitabine-based chemoresistance. Furthermore, antibody-mediated anti-TLR2 therapy suppressed the growth of human PDAC tumour xenografts, independent of a functional immune system. Our results support TLR2-based therapeutic targeting for precision medicine in PDAC, with further clinical utility that TLR2 activation is prognostic and predictive for chemoresponsiveness.
Collapse
Affiliation(s)
- Joanne Lundy
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Linden J Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Hugh Gao
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Clayton, VIC, Australia
| | - Alison C West
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Louise McLeod
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Virginie Deswaerte
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Liang Yu
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Sean Porazinski
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Marina Pajic
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Darlinghurst, NSW, Australia
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Daniel Croagh
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
- Department of Surgery (School of Clinical Sciences at Monash Health), Monash University, Clayton, VIC, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
12
|
Carpenter E, Nelson S, Bednar F, Cho C, Nathan H, Sahai V, di Magliano MP, Frankel TL. Immunotherapy for pancreatic ductal adenocarcinoma. J Surg Oncol 2021; 123:751-759. [PMID: 33595893 DOI: 10.1002/jso.26312] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/05/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a lethal cancer with an urgent need for better medical therapies. Efforts have been made to investigate the efficacy of immunotherapy, particularly given the hallmarks of immune suppression and exhaustion in PDAC tumors. Here, we review the molecular components responsible for the immune-privileged state in PDAC and provide an overview of the immunotherapeutic strategies for PDAC including vaccine therapy, checkpoint blockade, myeloid-targeted therapy, and immune agonist therapy.
Collapse
Affiliation(s)
- Eileen Carpenter
- Department of Internal Medicine, Division of Gastroenterology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah Nelson
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Filip Bednar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Clifford Cho
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Hari Nathan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Vaibhav Sahai
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | - Timothy L Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Javadrashid D, Baghbanzadeh A, Hemmat N, Hajiasgharzadeh K, Nourbakhsh NS, Lotfi Z, Baradaran B. Envisioning the immune system to determine its role in pancreatic ductal adenocarcinoma: Culprit or victim? Immunol Lett 2021; 232:48-59. [PMID: 33647329 DOI: 10.1016/j.imlet.2021.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/15/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma has a poor 5-year survival rate that makes it one of the most fatal human malignancies. Unfortunately, despite the serious improvement in the survival of most cancers, there has been a minor advance in pancreatic cancer (PC). Major advances in PC treatment have been assessed over the bygone twenty-year time span, yet some complications make the survival of the patients shorter. Getting to know the PC tumor microenvironment (TME) and the immunosuppression that happens during the pathogenesis of this malignancy could be a great help to understand the nature of the immune system and find better treatment modalities based on it. Although many immune cells are present in PC, immunosuppression of the TME leads to severe immune dysfunction in the patients, therefore immune effectors fail to do their functions. Lately, immunotherapy has been presented as one of the promising treatment strategies for different malignancies including hepatocellular carcinoma, melanoma, non-small cell lung cancer, and kidney cancer. In PC, there has been shown promising results centered around the TME, immune checkpoint inhibitors, cancer vaccines, and other approaches especially when used as combinational therapy. Here we dig a little deeper into the role of the immune system and possible therapeutic options in the treatment of PC.
Collapse
Affiliation(s)
- Darya Javadrashid
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Ziba Lotfi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Gnanasekaran J, Binder Gallimidi A, Saba E, Pandi K, Eli Berchoer L, Hermano E, Angabo S, Makkawi H, Khashan A, Daoud A, Elkin M, Nussbaum G. Intracellular Porphyromonas gingivalis Promotes the Tumorigenic Behavior of Pancreatic Carcinoma Cells. Cancers (Basel) 2020; 12:cancers12082331. [PMID: 32824786 PMCID: PMC7465784 DOI: 10.3390/cancers12082331] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/10/2020] [Accepted: 08/14/2020] [Indexed: 12/20/2022] Open
Abstract
Porphyromonas gingivalis is a member of the dysbiotic oral microbiome associated with oral inflammation and periodontal disease. Intriguingly, epidemiological studies link P. gingivalis to an increased risk of pancreatic cancer. Given that oral bacteria are detected in human pancreatic cancer, and both mouse and human pancreata harbor microbiota, we explored the involvement of P. gingivalis in pancreatic tumorigenesis using cell lines and a xenograft model. Live P. gingivalis induced proliferation of pancreatic cancer cells; however, surprisingly, this effect was independent of Toll-like receptor 2, the innate immune receptor that is engaged in response to P. gingivalis on other cancer and immune cells, and is required for P. gingivalis to induce alveolar bone resorption. Instead, we found that P. gingivalis survives inside pancreatic cancer cells, a trait that can be enhanced in vitro and is increased by hypoxia, a central characteristic of pancreatic cancer. Increased tumor cell proliferation was related to the degree of intracellular persistence, and infection of tumor cells with P. gingivalis led to enhanced growth in vivo. To the best of our knowledge, this study is the first to demonstrate the direct effect of exposure to P. gingivalis on the tumorigenic behavior of pancreatic cancer cell lines. Our findings shed light on potential mechanisms underlying the pancreatic cancer–periodontitis link.
Collapse
Affiliation(s)
- JebaMercy Gnanasekaran
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Adi Binder Gallimidi
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem 9112102, Israel;
| | - Elias Saba
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Karthikeyan Pandi
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Luba Eli Berchoer
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Esther Hermano
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem 9112102, Israel;
| | - Sarah Angabo
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Hasna′a Makkawi
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Arin Khashan
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Alaa Daoud
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
| | - Michael Elkin
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem 9112102, Israel;
- Correspondence: (M.E.); (G.N.); Tel.: +972-2-6776782 (M.E.); +972-2-6758581 (G.N.)
| | - Gabriel Nussbaum
- The Institute of Dental Sciences, Hebrew University, Hadassah Faculty of Dental Medicine, Jerusalem 9112102, Israel; (J.G.); (A.B.G.); (E.S.); (K.P.); (L.E.B.); (S.A.); (H.M.); (A.K.); (A.D.)
- Correspondence: (M.E.); (G.N.); Tel.: +972-2-6776782 (M.E.); +972-2-6758581 (G.N.)
| |
Collapse
|
15
|
Scheenstra MR, van Harten RM, Veldhuizen EJA, Haagsman HP, Coorens M. Cathelicidins Modulate TLR-Activation and Inflammation. Front Immunol 2020; 11:1137. [PMID: 32582207 PMCID: PMC7296178 DOI: 10.3389/fimmu.2020.01137] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Cathelicidins are short cationic peptides that are part of the innate immune system. At first, these peptides were studied mostly for their direct antimicrobial killing capacity, but nowadays they are more and more appreciated for their immunomodulatory functions. In this review, we will provide a comprehensive overview of the various effects cathelicidins have on the detection of damage- and microbe-associated molecular patterns, with a special focus on their effects on Toll-like receptor (TLR) activation. We review the available literature based on TLR ligand types, which can roughly be divided into lipidic ligands, such as LPS and lipoproteins, and nucleic-acid ligands, such as RNA and DNA. For both ligand types, we describe how direct cathelicidin-ligand interactions influence TLR activation, by for instance altering ligand stability, cellular uptake and receptor interaction. In addition, we will review the more indirect mechanisms by which cathelicidins affect downstream TLR-signaling. To place all this information in a broader context, we discuss how these cathelicidin-mediated effects can have an impact on how the host responds to infectious organisms as well as how these effects play a role in the exacerbation of inflammation in auto-immune diseases. Finally, we discuss how these immunomodulatory activities can be exploited in vaccine development and cancer therapies.
Collapse
Affiliation(s)
- Maaike R Scheenstra
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Roel M van Harten
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Edwin J A Veldhuizen
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Henk P Haagsman
- Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Utrecht University, Utrecht, Netherlands
| | - Maarten Coorens
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.,Department of Clinical Microbiology, Karolinska University Laboratory, Stockholm, Sweden
| |
Collapse
|
16
|
Patinote C, Karroum NB, Moarbess G, Cirnat N, Kassab I, Bonnet PA, Deleuze-Masquéfa C. Agonist and antagonist ligands of toll-like receptors 7 and 8: Ingenious tools for therapeutic purposes. Eur J Med Chem 2020; 193:112238. [PMID: 32203790 PMCID: PMC7173040 DOI: 10.1016/j.ejmech.2020.112238] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
The discovery of the TLRs family and more precisely its functions opened a variety of gates to modulate immunological host responses. TLRs 7/8 are located in the endosomal compartment and activate a specific signaling pathway in a MyD88-dependant manner. According to their involvement into various autoimmune, inflammatory and malignant diseases, researchers have designed diverse TLRs 7/8 ligands able to boost or block the inherent signal transduction. These modulators are often small synthetic compounds and most act as agonists and to a much lesser extent as antagonists. Some of them have reached preclinical and clinical trials, and only one has been approved by the FDA and EMA, imiquimod. The key to the success of these modulators probably lies in their combination with other therapies as recently demonstrated. We gather in this review more than 360 scientific publications, reviews and patents, relating the extensive work carried out by researchers on the design of TLRs 7/8 modulators, which are classified firstly by their biological activities (agonist or antagonist) and then by their chemical structures, which total syntheses are not discussed here. This review also reports about 90 clinical cases, thereby showing the biological interest of these modulators in multiple pathologies.
Collapse
Affiliation(s)
- Cindy Patinote
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France.
| | - Nour Bou Karroum
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France; Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Georges Moarbess
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | - Natalina Cirnat
- IBMM, Université de Montpellier, CNRS, ENSCM, Montpellier, France
| | - Issam Kassab
- Tumorigenèse et Pharmacologie Antitumorale, Lebanese University, EDST, BP 90656, Fanar Jdeideh, Lebanon
| | | | | |
Collapse
|
17
|
Das S, Shapiro B, Vucic EA, Vogt S, Bar-Sagi D. Tumor Cell-Derived IL1β Promotes Desmoplasia and Immune Suppression in Pancreatic Cancer. Cancer Res 2020; 80:1088-1101. [PMID: 31915130 PMCID: PMC7302116 DOI: 10.1158/0008-5472.can-19-2080] [Citation(s) in RCA: 214] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 11/22/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is an aggressive malignancy typified by a highly stromal and weakly immunogenic tumor microenvironment that promotes tumor evolution and contributes to therapeutic resistance. Here, we demonstrate that PDA tumor cell-derived proinflammatory cytokine IL1β is essential for the establishment of the protumorigenic PDA microenvironment. Tumor cell-derived IL1β promoted the activation and secretory phenotype of quiescent pancreatic stellate cells and established an immunosuppressive milieu mediated by M2 macrophages, myeloid-derived suppressor cells, CD1dhiCD5+ regulatory B cells, and Th17 cells. Loss of tumor cell-derived IL1 signaling in tumor stroma enabled intratumoral infiltration and activation of CD8+ cytotoxic T cells, attenuated growth of pancreatic neoplasia, and conferred survival advantage to PDA-bearing mice. Accordingly, antibody-mediated neutralization of IL1β significantly enhanced the antitumor activity of α-PD-1 and was accompanied by increased tumor infiltration of CD8+ T cells. Tumor cell expression of IL1β in vivo was driven by microbial-dependent activation of toll-like receptor 4 (TLR4) signaling and subsequent engagement of the NLRP3 inflammasome. Collectively, these findings identify a hitherto unappreciated role for tumor cell-derived IL1β in orchestrating an immune-modulatory program that supports pancreatic tumorigenesis. SIGNIFICANCE: These findings identify a new modality for immune evasion in PDA that depends on IL1β production by tumor cells through TLR4-NLRP3 inflammasome activation. Targeting this axis might provide an effective PDA therapeutic strategy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Carcinogenesis/immunology
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/immunology
- Carcinoma, Pancreatic Ductal/pathology
- Cell Line, Tumor/transplantation
- Disease Models, Animal
- Drug Synergism
- Epithelial Cells
- Female
- Humans
- Inflammasomes/immunology
- Inflammasomes/metabolism
- Interleukin-1beta/antagonists & inhibitors
- Interleukin-1beta/immunology
- Interleukin-1beta/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Transgenic
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Pancreatic Ducts/cytology
- Pancreatic Neoplasms/drug therapy
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/pathology
- Primary Cell Culture
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Signal Transduction/immunology
- Toll-Like Receptor 4/metabolism
- Tumor Escape/drug effects
- Tumor Escape/immunology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Shipra Das
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York
| | - Beny Shapiro
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York
| | - Emily A Vucic
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York
| | - Sandra Vogt
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York
| | - Dafna Bar-Sagi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York.
| |
Collapse
|
18
|
Goldberg R, Meirovitz A, Abecassis A, Hermano E, Rubinstein AM, Nahmias D, Grinshpun A, Peretz T, Elkin M. Regulation of Heparanase in Diabetes-Associated Pancreatic Carcinoma. Front Oncol 2019; 9:1405. [PMID: 31921662 PMCID: PMC6914686 DOI: 10.3389/fonc.2019.01405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
While at least six types of cancer have been associated with diabetes, pancreatic ductal adenocarcinoma (PDAC) and diabetes exhibit a unique bidirectional relationship. Recent reports indicate that majority of PDAC patients display hyperglycemia, and ~50% have concurrent diabetes. In turn, hyperglycemic/diabetic state in PDAC patients fosters enhanced growth and dissemination of the tumor. Heparanase enzyme (the sole mammalian endoglycosidase degrading glycosaminoglycan heparan sulfate) is tightly implicated in PDAC progression, aggressiveness, and therapy resistance. Overexpression of heparanase is a characteristic feature of PDAC, correlating with poor prognosis. However, given the lack of heparanase expression in normal pancreatic tissue, the regulatory mechanisms responsible for induction of the enzyme in PDAC have remained largely unknown. Previously reported inducibility of heparanase gene by diabetic milieu components in several non-cancerous cell types prompted us to hypothesize that in the setting of diabetes-associated PDAC, hyperglycemic state may induce heparanase overexpression. Here, utilizing a mouse model of diet-induced metabolic syndrome/diabetes, we found accelerated PDAC progression in hyperglycemic mice, occurring along with induction of heparanase in PDAC. In vitro, we demonstrated that advanced glycation end-products (AGE), which are largely thought as oxidative derivatives resulting from chronic hyperglycemia, and the receptor for AGE (RAGE) are responsible for heparanase induction in PDAC cells. These findings underscore the new mechanism underlying preferential expression of heparanase in pancreatic cancer. Moreover, taken together with the well-established causal role of the enzyme in PDAC progression, our findings indicate that heparanase may sustain (at least in part) reciprocal causality between diabetes and pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Rachel Goldberg
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alexia Abecassis
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Esther Hermano
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel M Rubinstein
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniela Nahmias
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Albert Grinshpun
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamar Peretz
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Michael Elkin
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
19
|
Hii LW, Lim SHE, Leong CO, Chin SY, Tan NP, Lai KS, Mai CW. The synergism of Clinacanthus nutans Lindau extracts with gemcitabine: downregulation of anti-apoptotic markers in squamous pancreatic ductal adenocarcinoma. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:257. [PMID: 31521140 PMCID: PMC6744713 DOI: 10.1186/s12906-019-2663-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 08/30/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Clinacanthus nutans extracts have been consumed by the cancer patients with the hope that the extracts can kill cancers more effectively than conventional chemotherapies. Our previous study reported its anti-inflammatory effects were caused by inhibiting Toll-like Receptor-4 (TLR-4) activation. However, we are unsure of its anticancer effect, and its interaction with existing chemotherapy. METHODS We investigated the anti-proliferative efficacy of polar leaf extracts (LP), non-polar leaf extracts (LN), polar stem extract (SP) and non-polar stem extracts (SN) in human breast, colorectal, lung, endometrial, nasopharyngeal, and pancreatic cancer cells using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, MTT assay. The most potent extracts was tested along with gemcitabine using our established drug combination analysis. The effect of the combinatory treatment in apoptosis were quantified using enzyme-linked immunosorbent assay (ELISA), Annexin V assay, antibody array and immunoblotting. Statistical significance was analysed using one-way analysis of variance (ANOVA) and post hoc Dunnett's test. A p-value of less than 0.05 (p < 0.05) was considered statistical significance. RESULTS All extracts tested were not able to induce potent anti-proliferative effects. However, it was found that pancreatic ductal adenocarcinoma, PDAC (AsPC1, BxPC3 and SW1990) were the cell lines most sensitive cell lines to SN extracts. This is the first report of C. nutans SN extracts acting in synergy with gemcitabine, the first line chemotherapy for pancreatic cancer, as compared to conventional monotherapy. In the presence of SN extracts, we can reduce the dose of gemcitabine 2.38-5.28 folds but still maintain the effects of gemcitabine in PDAC. SN extracts potentiated the killing of gemcitabine in PDAC by apoptosis. Bax was upregulated while bcl-2, cIAP-2, and XIAP levels were downregulated in SW1990 and BxPC3 cells treated with gemcitabine and SN extracts. The synergism was independent of TLR-4 expression in pancreatic cancer cells. CONCLUSION These results provide strong evidence of C. nutans extracts being inefficacious as monotherapy for cancer. Hence, it should not be used as a total substitution for any chemotherapy agents. However, SN extracts may synergise with gemcitabine in the anti-tumor mechanism.
Collapse
Affiliation(s)
- Ling-Wei Hii
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000 Malaysia
| | - Swee-Hua Erin Lim
- Perdana University-Royal College of Surgeons in Ireland, Seri Kembangan, 43400 Selangor Malaysia
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi, United Arab Emirates
| | - Chee-Onn Leong
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, 57000 Malaysia
- Centre for Cancer and Stem Cells Research, Institute for Research Development and Innovation, International Medical University, Kuala Lumpur, 57000 Malaysia
| | - Swee-Yee Chin
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Ngai-Paing Tan
- Department of Land Management, Faculty of Agriculture, Universiti Putra Malaysia, Seri Kembangan, 43400 Selangor Malaysia
| | - Kok-Song Lai
- Health Sciences Division, Abu Dhabi Women’s College, Higher Colleges of Technology, Abu Dhabi, United Arab Emirates
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400 Seri Kembangan, Selangor Malaysia
| | - Chun-Wai Mai
- Centre for Cancer and Stem Cells Research, Institute for Research Development and Innovation, International Medical University, Kuala Lumpur, 57000 Malaysia
- Department of Pharmaceutical Chemistry, School of Pharmacy, International Medical University, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Taskin E, Guven C, Kaya ST, Sahin L, Kocahan S, Degirmencioglu AZ, Gur FM, Sevgiler Y. The role of toll-like receptors in the protective effect of melatonin against doxorubicin-induced pancreatic beta cell toxicity. Life Sci 2019; 233:116704. [DOI: 10.1016/j.lfs.2019.116704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 02/08/2023]
|
21
|
Domenis R, Cifù A, Marinò D, Fabris M, Niazi KR, Soon-Shiong P, Curcio F. Toll-like Receptor-4 Activation Boosts the Immunosuppressive Properties of Tumor Cells-derived Exosomes. Sci Rep 2019; 9:8457. [PMID: 31186484 PMCID: PMC6560033 DOI: 10.1038/s41598-019-44949-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 05/24/2019] [Indexed: 12/28/2022] Open
Abstract
The biology of tumor-derived exosomes (TEX) is only partially understood and much remains to be studied in order to define the effect that the tumor microenvironment or the activation of tumor cells exerts on their composition and functions. Increased expression and activity of toll-like receptor 4 (TLR4) in chronic infectious and inflammatory conditions is related with cancer progression: its activation induces an inflammatory signaling that increases the tumorigenic potential of cancer cells promoting their immune evasion. We investigated the immune modulatory properties of TEX released upon cell TLR4 activation, and we found that, although differences were observed depending on the type of the tumor, the treatment influences TEX composition and boosts their immunosuppressive ability. Our results suggest that the activation of TLR4 supports tumor progression by stimulating the release of more effective immunosuppressive exosomes, which allow tumor cells to escape immune surveillance and probably even play a role in the metastatic process.
Collapse
Affiliation(s)
- Rossana Domenis
- Dipartimento di Area Medica (DAME), Università degli Studi di Udine, Udine, Italy
| | - Adriana Cifù
- Dipartimento di Area Medica (DAME), Università degli Studi di Udine, Udine, Italy
| | - Daniele Marinò
- Dipartimento di Area Medica (DAME), Università degli Studi di Udine, Udine, Italy
| | - Martina Fabris
- Dipartimento di Area Medica (DAME), Università degli Studi di Udine, Udine, Italy.,Istituto di Patologia Clinica, Azienda Sanitaria Universitaria Integrata di Udine (ASUID), Udine, Italy
| | | | | | - Francesco Curcio
- Dipartimento di Area Medica (DAME), Università degli Studi di Udine, Udine, Italy. .,Istituto di Patologia Clinica, Azienda Sanitaria Universitaria Integrata di Udine (ASUID), Udine, Italy.
| |
Collapse
|
22
|
Panebianco C, Pazienza V. Body site-dependent variations of microbiota in pancreatic cancer pathophysiology. Crit Rev Clin Lab Sci 2019; 56:260-273. [PMID: 31060399 DOI: 10.1080/10408363.2019.1615407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Lack of specific symptoms and reliable biomarkers, along with aggressive nature and resistance to therapies makes pancreatic cancer (PC) one of the leading causes of death from cancer worldwide. The search for new diagnostic, prognostic, predictive, and therapeutic tools that could improve clinical outcomes of patients has led, in recent years, to the investigation of potential roles for the microbiota in the pathogenesis of this disease. The human microbiota encompasses trillions of microorganisms residing within several body tissues and organs, where they provide beneficial functions for host homeostasis and health. Derangements of the microbial ecology in different anatomic districts have been described in PC, as in many other diseases, both in patients and in animal models. In detail, infection from the gastric pathogen Helicobacter pylori and changes in composition and diversity of oral, intestinal, and pancreatic microbiota have been found to associate with PC. Future research should assess how to potentially exploit such differences in microbiota composition as diagnostic, prognostic, or predictive biomarkers, and as targets for therapeutic interventions, in the hope of improving the dismal prognosis of this insidious cancer.
Collapse
Affiliation(s)
- Concetta Panebianco
- a Division of Gastroenterology , Fondazione IRCCS Casa Sollievo della Sofferenza , San Giovanni Rotondo , Italy
| | - Valerio Pazienza
- a Division of Gastroenterology , Fondazione IRCCS Casa Sollievo della Sofferenza , San Giovanni Rotondo , Italy
| |
Collapse
|
23
|
Chen X, Cheng F, Liu Y, Zhang L, Song L, Cai X, You T, Fan X, Wang D, Gong A, Zhu H. Toll-like receptor 2 and Toll-like receptor 4 exhibit distinct regulation of cancer cell stemness mediated by cell death-induced high-mobility group box 1. EBioMedicine 2019; 40:135-150. [PMID: 30679086 PMCID: PMC6413584 DOI: 10.1016/j.ebiom.2018.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/26/2018] [Accepted: 12/07/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND High-mobility group box 1 (HMGB1), a common extracellular damage associated molecular pattern molecule, is overexpressed in several solid tumors including pancreatic carcinoma. We previously observed that radiotherapy induced dying cells secrete HMGB1 and accelerate pancreatic carcinoma progression through an unclear mechanism. METHODS Using the Millicell system as an in vitro co-culture model, we performed quantitative reverse transcriptase-polymerase chain reaction, western blot and sphere forming ability analyses to access the effect of dying-cell-derived HMGB1 on CD133+ cancer cell stemness in vitro and in vivo. Interactions between HMGB1 and Toll-like receptor 2(TLR2)/TLR4 were studied by co- immunoprecipitation. Western blot and short-hairpin RNA-based knockdown assays were conducted to detect HMGB1 and TLR2/TLR4 signaling activity. FINDINGS Radiation-associated, dying-cell-derived HMGB1 maintained stemness and contributed to CD133+ cancer stem cell self-renewal in vitro and in vivo. In overexpressing and silencing experiments, we demonstrated that the process was activated by TLR2 receptor, whereas TLR4 antagonized HMGB1-TLR2 signaling. Wnt/β-catenin signaling supported the HMGB1-TLR2 mediated stemness of CD133+ cancer cells. INTERPRETATION Our results show how irradiation-induced cell death might enhance the stemness of resident cancer cells, and indicate HMGB1-TLR2 signaling as a potential therapeutic target for preventing pancreatic cancer recurrence.
Collapse
Affiliation(s)
- Xuelian Chen
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Fang Cheng
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- School of Pharmaceutical Sciences (Shenzhen), SYSU, 510006, China
- Faculty of Science and Engineering, Åbo Akademi University and Turku Centre for Biotechnology, Turku FI-20520, Finland
| | - Yanfang Liu
- Department of Central Laboratory, The First People's Hospital of Zhenjiang, Zhenjiang 212001, China
| | - Lirong Zhang
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Lian Song
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xiaojie Cai
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Tao You
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Xin Fan
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Dongqing Wang
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Haitao Zhu
- Central laboratory of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| |
Collapse
|
24
|
Cai Y, Huang J, Xing H, Li B, Li L, Wang X, Peng D, Chen J. Contribution of FPR and TLR9 to hypoxia-induced chemoresistance of ovarian cancer cells. Onco Targets Ther 2018; 12:291-301. [PMID: 30643427 PMCID: PMC6314315 DOI: 10.2147/ott.s190118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background/purpose The aim of this study was to investigate the role and mechanisms of the formyl peptide receptor (FPR) and the toll-like receptor 9 (TLR9) in hypoxia-induced chemoresistance of human ovarian cancer cells. Materials and methods SKOV3 cells were exposed to hypoxia for 24 hours, the supernatant was collected to stimulate normoxia-cultured SKOV3, and the inhibition rate of cell growth was detected with CCK8 test. The agonist of TLR9 CpG ODN and the agonist of FPR fMLF were applied to investigate the chemosensitivity of SKOV3 cells to cisplatin. The cells were also treated with FPR antagonist t-Boc or TLR9 antagonist CQ. Western blot was applied to detect protein levels of FPR, TLR9, MRP, P-gp, p53 and Beclin-1. Immunofluorescence staining was applied to observe the distribution of TLR9 in SKOV3 cells. Results Hypoxia exposure reduced the inhibition rate of cisplatin on SKOV3 cells. WB showed that FPR and TLR9 were expressed in human ovarian cancer tissues and SKOV3 cells, and the levels were increased with longer hypoxia time. After SKOV3 was stimulated with fMLF or ODN2006, cisplatin-induced inhibition rate was significantly decreased. tBoc and CQ significantly attenuated hypoxia supernatant-induced chemoresistance of SKOV3 cells. Hypoxia supernatants significantly increased MRP, P-gp, p53 and Beclin-1 proteins in SKOV3 cells, which were significantly reduced by tBoc. Conclusion Hypoxia upregulates the expression of FPR and TLR9, and promotes the release of ligands for both receptors in human ovarian cancer cell line. FPR and TLR9 may be noval targets for chemosensitizing to ovarian cancer cells.
Collapse
Affiliation(s)
- Yongqing Cai
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Jian Huang
- Department of High Altitude Biology and Pathology, High Altitude Military Medical College, Army Medical University, Chongqing 400042, China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Bin Li
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Ling Li
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Xianfeng Wang
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Dan Peng
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital and Research Institute of Surgery, Army Medical University, Chongqing 400042, China,
| |
Collapse
|
25
|
Leppänen J, Helminen O, Huhta H, Kauppila JH, Isohookana J, Haapasaari KM, Karihtala P, Parkkila S, Saarnio J, Lehenkari PP, Karttunen TJ. Toll-like receptors 2, 4 and 9 and hypoxia markers HIF-1alpha and CAIX in pancreatic intraepithelial neoplasia. APMIS 2018; 126:852-863. [PMID: 30357962 DOI: 10.1111/apm.12894] [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: 05/07/2018] [Accepted: 09/13/2018] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer arises from precursor lesions called pancreatic intraepithelial neoplasia (PanIN) characterized by inflammatory microenvironment. In pancreatic cancer, strong innate immunity and hypoxia responses are typical. Occurrence and relationship of these responses in human PanINs is unknown. We have studied the expression of toll-like receptors (TLR) TLR2, TLR4 and TLR9, and hypoxia markers HIF-1alpha and Carbonic anhydrase IX (CAIX) in normal and inflamed pancreatic ducts, in PanINs and in cancers. The samples of 69 surgically resected pancreatic ductal adenocarcinoma patients were stained using immunohistochemistry. We found TLR2, TLR9, HIF-1alpha and CAIX to be prominently expressed in pancreatic intraepithelial neoplasia. Expression of TLR2 showed a linear increase from PanIN1 to PanIN3, while the highest TLR4 expression was detected in inflamed ducts, and TLR9 expression in PanIN1 lesions. Within the PanIN1-group, nuclear HIF-1alpha correlated with membranous and cytoplasmic TLR2 expression (ρ = 0.982 and 0.815; p < 0.001 and p = 0.025, respectively), and in the PanIN2-group nuclear HIF-1alpha correlated with nuclear TLR9 expression 0.636, p = 0.026). Our findings show that the expression of TLRs 2, 4 and 9, and hypoxia markers HIF-1alpha and CAIX is abnormal in pancreatic intraepithelial neoplasia suggesting that both the innate immunity activation and hypoxia response are involved in early pancreatic carcinogenesis. However, these processes might be independent.
Collapse
Affiliation(s)
- Joni Leppänen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Olli Helminen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Heikki Huhta
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Joonas H Kauppila
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland.,Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Joel Isohookana
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Kirsi-Maria Haapasaari
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Peeter Karihtala
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Seppo Parkkila
- School of Medicine, University of Tampere, Tampere, Finland.,Fimlab Ltd., Tampere University Hospital, Tampere, Finland
| | - Juha Saarnio
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Petri P Lehenkari
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Tuomo J Karttunen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| |
Collapse
|
26
|
Oral Health and the Oral Microbiome in Pancreatic Cancer: An Overview of Epidemiological Studies. ACTA ACUST UNITED AC 2018; 23:310-314. [PMID: 29189325 DOI: 10.1097/ppo.0000000000000287] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE The aim was to provide a cohesive overview of epidemiological studies of periodontal disease, oral microbiome profiles, and pancreatic cancer risk. DESIGN A PubMed search of articles published in English through July 2017 with additional review of bibliographies of identified articles. RESULTS Risk estimates for periodontal disease associated with pancreatic cancer consistently ranged from 1.5 to 2, aligning with a meta-analysis summary relative risk of 1.74. Analyses of antibodies to pathogenic and/or commensal oral bacteria in prediagnostic blood provided evidence that some oral bacteria and oral microbial diversity may be related to pancreatic cancer. CONCLUSIONS Overall, the data present a plausible but complex relationship among pancreatic cancer, the oral microbiome, periodontal disease, and other risk factors that might be explained by systemic effects on immune and inflammatory processes. Larger comprehensive studies that examine serially collected epidemiological/clinical data and blood, tissue, and various microbial samples are needed to definitively determine how and whether oral health-related factors contribute to pancreatic cancer risk.
Collapse
|
27
|
Lin R, Wang Y, Chen Q, Liu Z, Xiao S, Wang B, Shi B. TRPM2 promotes the proliferation and invasion of pancreatic ductal adenocarcinoma. Mol Med Rep 2018; 17:7537-7544. [PMID: 29620272 PMCID: PMC5983953 DOI: 10.3892/mmr.2018.8816] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 01/29/2018] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate transient receptor potential cation channel subfamily M member 2 (TRPM2), a promising therapeutic target and biomarker for pancreatic ductal adenocarcinoma (PDAC) prognosis, in addition to determining its effects regarding tumor progression and invasion. PDAC is a fatal disease with a poor prognosis, and its associated pathogenic molecular mechanisms remain to be determined. In the present study, combined analysis using genomic and transcriptomic data from two PDAC studies was performed to discover a survival‑associated biomarker of PDAC. Survival analysis for genes mutated in ≥10 patients was performed using a Kaplan‑Meier curve and tested for significance using a log‑rank test. Furthermore, gene‑expression correlation analysis was performed to determine the genes with the strongest correlations to TRPM2. In addition, a Cell Counting Kit‑8 assay, a scratch wound‑healing assay and a Transwell assay were performed in the present study to investigate the proliferative, invasive and metastatic ability of PANC‑1 cells in TRPM2‑overexpressing and downregulated groups. The mutated TRPM2 gene had a strong negative correlation with patient survival probability compared with the normal control group (P=1.06x10‑4). Expression of TRPM2 was strongly correlated with expression of probable phospholipid‑transporting ATPase IM, γ‑parvin, tudor domain containing 9, Toll‑like receptor 7 and Scm‑like with four MBT domains protein 2 according to the criterion of a correlation coefficient >0.5. Furthermore, the results of the present study demonstrated that the TRPM2 overexpression in a PDAC cell line (PANC‑1) promoted cell proliferation, invasion and metastatic ability. TRPM2 represents a potential therapeutic target and prognostic marker for patients with PDAC. TRPM2 regulates cell proliferation, invasion and migration; however, the underlying mechanism requires further investigation in future studies.
Collapse
Affiliation(s)
- Rui Lin
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Yufeng Wang
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Quanning Chen
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Zhongyan Liu
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Shuai Xiao
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Bingyi Wang
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| | - Baomin Shi
- Department of General Surgery, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, P.R. China
| |
Collapse
|
28
|
Neoptolemos JP, Kleeff J, Michl P, Costello E, Greenhalf W, Palmer DH. Therapeutic developments in pancreatic cancer: current and future perspectives. Nat Rev Gastroenterol Hepatol 2018; 15:333-348. [PMID: 29717230 DOI: 10.1038/s41575-018-0005-x] [Citation(s) in RCA: 744] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The overall 5-year survival for pancreatic cancer has changed little over the past few decades, and pancreatic cancer is predicted to be the second leading cause of cancer-related mortality in the next decade in Western countries. The past few years, however, have seen improvements in first-line and second-line palliative therapies and considerable progress in increasing survival with adjuvant treatment. The use of biomarkers to help define treatment and the potential of neoadjuvant therapies also offer opportunities to improve outcomes. This Review brings together information on achievements to date, what is working currently and where successes are likely to be achieved in the future. Furthermore, we address the questions of how we should approach the development of pancreatic cancer treatments, including those for patients with metastatic, locally advanced and borderline resectable pancreatic cancer, as well as for patients with resected tumours. In addition to embracing newer strategies comprising genomics, stromal therapies and immunotherapies, conventional approaches using chemotherapy and radiotherapy still offer considerable prospects for greater traction and synergy with evolving concepts.
Collapse
Affiliation(s)
- John P Neoptolemos
- Department of General Surgery, University of Heidelberg, Heidelberg, Germany.
| | - Jörg Kleeff
- Department of Visceral, Vascular and Endocrine Surgery, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany. .,Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.
| | - Patrick Michl
- Department of Internal Medicine I, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Eithne Costello
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - William Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Daniel H Palmer
- Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
29
|
Rajendrakumar SK, Uthaman S, Cho CS, Park IK. Nanoparticle-Based Phototriggered Cancer Immunotherapy and Its Domino Effect in the Tumor Microenvironment. Biomacromolecules 2018; 19:1869-1887. [DOI: 10.1021/acs.biomac.8b00460] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Santhosh Kalash Rajendrakumar
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, South Korea
| | - Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, South Korea
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul 08826, South Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University, Chonnam National University Medical School, Gwangju 61469, South Korea
| |
Collapse
|
30
|
Johnson BA, Yarchoan M, Lee V, Laheru DA, Jaffee EM. Strategies for Increasing Pancreatic Tumor Immunogenicity. Clin Cancer Res 2018; 23:1656-1669. [PMID: 28373364 DOI: 10.1158/1078-0432.ccr-16-2318] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/23/2017] [Accepted: 01/27/2017] [Indexed: 12/15/2022]
Abstract
Immunotherapy has changed the standard of care for multiple deadly cancers, including lung, head and neck, gastric, and some colorectal cancers. However, single-agent immunotherapy has had little effect in pancreatic ductal adenocarcinoma (PDAC). Increasing evidence suggests that the PDAC microenvironment is comprised of an intricate network of signals between immune cells, PDAC cells, and stroma, resulting in an immunosuppressive environment resistant to single-agent immunotherapies. In this review, we discuss differences between immunotherapy-sensitive cancers and PDAC, the complex interactions between PDAC stroma and suppressive tumor-infiltrating cells that facilitate PDAC development and progression, the immunologic targets within these complex networks that are druggable, and data supporting combination drug approaches that modulate multiple PDAC signals, which should lead to improved clinical outcomes. Clin Cancer Res; 23(7); 1656-69. ©2017 AACRSee all articles in this CCR Focus section, "Pancreatic Cancer: Challenge and Inspiration."
Collapse
Affiliation(s)
- Burles A Johnson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Mark Yarchoan
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Valerie Lee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Daniel A Laheru
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland. .,Department of Pathology, Sidney Kimmel Comprehensive Cancer Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland
| |
Collapse
|
31
|
Ding SM, Lu AL, Zhang W, Zhou L, Xie HY, Zheng SS, Li QY. The role of cancer-associated fibroblast MRC-5 in pancreatic cancer. J Cancer 2018; 9:614-628. [PMID: 29483967 PMCID: PMC5820929 DOI: 10.7150/jca.19614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 11/25/2017] [Indexed: 02/06/2023] Open
Abstract
Background: Our previous study showed that cancer-associated fibroblast MRC-5 promoted hepatocellular carcinoma progression by enhancing migration and invasion capability. However, few studies have explored the role of MRC-5 in pancreatic cancer (PC). In this study, we examined the exact role and associated mechanisms of MRC-5. Methods: The conditioned media for MRC-5 was used to culture PC cell lines SW1990 and PANC-1. Cell proliferation was compared based on colony formation assays of PC cells in normal media and of PC cells cultured with conditioned media of MRC-5. Cell migration and invasion were assayed by transwell chambers. The expression of EMT-related proteins and apoptosis-related proteins was evaluated using Western blot. And confocal microscopy was used to further detect the expression of EMT-related proteins. qRT-PCR was used to confirm the expression changes of related genes at the mRNA level. We also used flow cytometry to examine the cell cycle, apoptotic rate, and expression of CD3, CD4, CD14, CD25, CD45, CD61, CD90, TLR1, and TLR4. Results: MRC-5 repressed the colony formation ability of PC cells and significantly inhibited cell migration and invasion potential. MRC-5 induced S-phase cell cycle arrest but did not augment the apoptotic effects in PC cells. We hypothesized that the weakened malignant biological behavior of PC cells was correlated with MRC-5-induced altered expression of the cancer stem cell marker CD90; the immune-related cell surface molecules CD14, CD25, TLR4, and TLR1; and cell polarity complexes Par, Scribble, and Crumbs. Conclusion: MRC-5 limits the malignant activities of PC cells by suppressing cancer stem cell expansion, remolding epithelial polarity, and blocking the protumoral cascade reaction coupled to TLR4, TLR1, CD14, and CD25.
Collapse
Affiliation(s)
- Song-Ming Ding
- Shulan (Hangzhou) Hospital (Zhejiang University International Hospital), Hangzhou, Zhejiang, P.R. China
| | - Ai-Li Lu
- Division of oncology department, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Wu Zhang
- Shulan (Hangzhou) Hospital (Zhejiang University International Hospital), Hangzhou, Zhejiang, P.R. China
| | - Lin Zhou
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Trans-plantation, Zhejiang Province; Hangzhou, Zhejiang, China
| | - Hai-Yang Xie
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Trans-plantation, Zhejiang Province; Hangzhou, Zhejiang, China
| | - Shu-Sen Zheng
- Shulan (Hangzhou) Hospital (Zhejiang University International Hospital), Hangzhou, Zhejiang, P.R. China
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health; Key Laboratory of Organ Trans-plantation, Zhejiang Province; Hangzhou, Zhejiang, China
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Qi-Yong Li
- Shulan (Hangzhou) Hospital (Zhejiang University International Hospital), Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
32
|
Yan Y, Lu B, Li P, Wang J. NOD receptor and TLR9 modulation in severe acute pancreatitis‑induced intestinal injury. Mol Med Rep 2017; 16:8471-8476. [PMID: 28990073 DOI: 10.3892/mmr.2017.7661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 05/09/2017] [Indexed: 11/05/2022] Open
Abstract
Severe acute pancreatitis (SAP) has a rapid onset and may cause multiple organ dysfunction syndrome (MODS), which has high mortality. Nucleotide binding oligomerization domain (NOD) receptor and Toll‑like receptor 9 (TLR9), a pattern recognition receptor in innate immunity, are involved in inflammation, immunity and pathogen recognition. The role and mechanism of the NOD receptor and TLR9 in early MODS of SAP‑induced intestinal injury, however, remain unclear. Wistar rats were divided into control, SAP, TLR9 inhibitor and NOD receptor activation groups. Reverse transcription‑quantitative polymerase chain reaction was used to analyze the expression of TLR9, NOD1 and NOD2 in the experimental treatment groups. Serum amylase, creatinine and alanine aminotransferase indices were measured, ELISA was used to determine the expression of tumor necrosis factor‑α (TNF‑α) and interleukin‑1β (IL‑1β) and western blot analysis was used to assess nuclear factor (NF)‑κB expression levels in intestinal tissues. Reactive oxygen species (ROS) levels and superoxide dismutase (SOD) activity were quantified by spectrometry. SAP and NOD receptor activation groups exhibited significantly elevated TLR9, NOD1, NOD2, TNF‑α, IL‑1β and nuclear factor (NF)‑κB levels compared with the control group. Furthermore, ROS production was increased, SOD activity was decreased and higher serum indices were exhibited, compared with the control group. The NOD receptor group presented more significant differences compared with the SAP group. The TLR9 inhibitor group exhibited opposite effects, with markedly decreased TLR9, NOD1, NOD2, TNF‑α, IL‑1β and NF‑κB levels. The TLR9 inhibitor group also presented reduced ROS production, increased SOD activity and lower serum indexes compared to the SAP group. The present study therefore indicated that NOD receptor and TLR9 may modulate the inflammatory response and further impact upon intestinal injury of SAP, via the regulation of NF‑κB expression and the oxidation/antioxidation balance, suggesting therapeutically targeting NOD receptor and TLR9 might be a useful approach for the treatment of severe acute pancreatitis.
Collapse
Affiliation(s)
- Yupeng Yan
- Intensive Care Unit, China Meitan General Hospital, Beijing 100028, P.R. China
| | - Bin Lu
- Department of Anesthesiology, Beijing Hospital of Traditional Chinese Medicine, Beijing 100010, P.R. China
| | - Pengyang Li
- Department of Orthopedics, Dongzhimen Hospital, Beijing 100010, P.R. China
| | - Ji Wang
- Intensive Care Unit, China Meitan General Hospital, Beijing 100028, P.R. China
| |
Collapse
|
33
|
Beyer K, Partecke LI, Roetz F, Fluhr H, Weiss FU, Heidecke CD, von Bernstorff W. LPS promotes resistance to TRAIL-induced apoptosis in pancreatic cancer. Infect Agent Cancer 2017; 12:30. [PMID: 28572836 PMCID: PMC5450120 DOI: 10.1186/s13027-017-0139-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 05/16/2017] [Indexed: 12/29/2022] Open
Abstract
Background Though TRAIL has been hailed as a promising drug for tumour treatment, it has been observed that many tumour cells have developed escape mechanisms against TRAIL-induced apoptosis. As a receptor of LPS, TLR 4, which is expressed on a variety of cancer cells, can be associated with TRAIL-resistance of tumour cells and tumour progression as well as with the generation of an anti-tumour immune response. Methods In this study, the sensitivity to TRAIL-induced apoptosis as well as the influence of LPS-co-stimulation on the cell viability of the pancreatic cancer cell lines PANC-1, BxPC-3 and COLO 357 was examined by FACS analyses and a cell viability assay. Subsequently, the expression of TRAIL-receptors was detected via FACS analyses. Levels of osteoprotegerin (OPG) were also determined using an enzyme-linked immunosorbent assay. Results PANC-1 cells were shown to be resistant to TRAIL-induced apoptosis. This was accompanied by significantly increased osteoprotegerin levels and a significantly decreased expression of DR4. In contrast, TRAIL significantly induced apoptosis in COLO 357 cells and to a lesser degree in BxPC-3 cells. Co-stimulation of COLO 357 as well as BxPC-3 cells combining TRAIL and LPS resulted in a significant decrease in TRAIL-induced apoptosis. In COLO 357 cells TRAIL-stimulation decreased the levels of OPG thereby not altering the expression of the TRAIL-receptors 1–4 resulting in a high susceptibility to TRAIL-induced apoptosis. Co-stimulation with LPS and TRAIL completely reversed the effect of TRAIL on OPG levels reaching a 2-fold increase beyond the level of non-stimulated cells resulting in a lower susceptibility to apoptosis. In BxPC-3, TRAIL stimulation decreased the expression of DR4 and significantly increased the decoy receptors TRAIL-R3 and TRAIL-R4 leading to a decrease in TRAIL-induced apoptosis. OPG levels remained unchanged. Co-stimulation with TRAIL and LPS further enhanced the changes in TRAIL-receptor-expression promoting apoptosis resistance. Conclusions Here it has been shown that TRAIL-resistance in pancreatic cancer cells can be mediated by the inflammatory molecule LPS as well as by different expression patterns of functional and non-functional TRAIL-receptors.
Collapse
Affiliation(s)
- Katharina Beyer
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany.,Department of General, Visceral and Vascular Surgery, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Ivo Partecke
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Felicitas Roetz
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Herbert Fluhr
- Department of Medicine A, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Frank Ulrich Weiss
- Department of Obstetrics and Gynaecology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - Claus-Dieter Heidecke
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| | - Wolfram von Bernstorff
- Department of General, Visceral, Thoracic and Vascular Surgery, Universitätsmedizin Greifswald, Greifswald, Germany
| |
Collapse
|
34
|
Leppänen J, Helminen O, Huhta H, Kauppila JH, Isohookana J, Haapasaari KM, Lehenkari P, Saarnio J, Karttunen TJ. High toll-like receptor (TLR) 9 expression is associated with better prognosis in surgically treated pancreatic cancer patients. Virchows Arch 2017; 470:401-410. [PMID: 28191612 DOI: 10.1007/s00428-017-2087-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/10/2017] [Accepted: 02/02/2017] [Indexed: 12/14/2022]
Abstract
Pancreatic cancer remains one of the deadliest malignancies in the world. Inflammatory response and tumor environment are thought to play a major role in its pathogenesis. Knowledge on TLR expression and impact on patient survival in pancreatic cancer is limited. The study's aim was to clarify the role of different TLRs in pancreatic cancer. TLR2, TLR4, and TLR9 expression was investigated in 65 surgically resected pancreatic ductal adenocarcinoma specimens by immunohistochemistry. The association between TLR expression, clinical parameters, and local inflammatory response to the tumor was assessed using chi-square test. Relation between patient survival and TLR expression was calculated with multivariable Cox regression, adjusted for age, sex, and tumor stage. We found TLR2, TLR4, and TLR9 to be expressed in pancreatic cancer. There was no association between TLR expression and tumor stage, tumor size, lymph node metastasis, or tumor necrosis. Contrary to our initial hypothesis, high cytoplasmic TLR9 expression was associated with longer patient survival, and multivariate analysis identified low TLR9 expression as an independent risk factor for cancer-specific death (HR 3.090, 95% CI 1.673-5.706). The results suggest that high TLR9 expression in pancreatic ductal adenocarcinoma indicates improved prognosis. The prognostic effect of TLR9 might be associated with bacterial exposure, but this needs further evidence.
Collapse
Affiliation(s)
- Joni Leppänen
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland.
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland.
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland.
| | - Olli Helminen
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| | - Heikki Huhta
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| | - Joonas H Kauppila
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, 17176, Stockholm, Sweden
| | - Joel Isohookana
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| | - Kirsi-Maria Haapasaari
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| | - Petri Lehenkari
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
- Department of Anatomy and Cell Biology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
| | - Juha Saarnio
- Department of Surgery, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| | - Tuomo J Karttunen
- Department of Pathology, University of Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Medical Research Center Oulu, P.O. Box 5000, 90014, Oulu, Finland
- Oulu University Hospital, P.O. Box 21, 90029, Oulu, Finland
| |
Collapse
|
35
|
Binker-Cosen MJ, Richards D, Oliver B, Gaisano HY, Binker MG, Cosen-Binker LI. Palmitic acid increases invasiveness of pancreatic cancer cells AsPC-1 through TLR4/ROS/NF-κB/MMP-9 signaling pathway. Biochem Biophys Res Commun 2017; 484:152-158. [DOI: 10.1016/j.bbrc.2017.01.051] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 02/07/2023]
|
36
|
Grimmig T, Moench R, Kreckel J, Haack S, Rueckert F, Rehder R, Tripathi S, Ribas C, Chandraker A, Germer CT, Gasser M, Waaga-Gasser AM. Toll Like Receptor 2, 4, and 9 Signaling Promotes Autoregulative Tumor Cell Growth and VEGF/PDGF Expression in Human Pancreatic Cancer. Int J Mol Sci 2016; 17:ijms17122060. [PMID: 27941651 PMCID: PMC5187860 DOI: 10.3390/ijms17122060] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 11/23/2016] [Accepted: 12/02/2016] [Indexed: 02/07/2023] Open
Abstract
Toll like receptor (TLR) signaling has been suggested to play an important role in the inflammatory microenvironment of solid tumors and through this inflammation-mediated tumor growth. Here, we studied the role of tumor cells in their process of self-maintaining TLR expression independent of inflammatory cells and cytokine milieu for autoregulative tumor growth signaling in pancreatic cancer. We analyzed the expression of TLR2, -4, and -9 in primary human cancers and their impact on tumor growth via induced activation in several established pancreatic cancers. TLR-stimulated pancreatic cancer cells were specifically investigated for activated signaling pathways of VEGF/PDGF and anti-apoptotic Bcl-xL expression as well as tumor cell growth. The primary pancreatic cancers and cell lines expressed TLR2, -4, and -9. TLR-specific stimulation resulted in activated MAP-kinase signaling, most likely via autoregulative stimulation of demonstrated TLR-induced VEGF and PDGF expression. Moreover, TLR activation prompted the expression of Bcl-xL and has been demonstrated for the first time to induce tumor cell proliferation in pancreatic cancer. These findings strongly suggest that pancreatic cancer cells use specific Toll like receptor signaling to promote tumor cell proliferation and emphasize the particular role of TLR2, -4, and -9 in this autoregulative process of tumor cell activation and proliferation in pancreatic cancer.
Collapse
Affiliation(s)
- Tanja Grimmig
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Romana Moench
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Jennifer Kreckel
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Stephanie Haack
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Felix Rueckert
- Surgical Clinic Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| | - Roberta Rehder
- Medical School, Evangelic Faculty of Paraná, 80730-000 Curitiba, Brazil.
| | - Sudipta Tripathi
- Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Carmen Ribas
- Medical School, Evangelic Faculty of Paraná, 80730-000 Curitiba, Brazil.
| | - Anil Chandraker
- Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Christoph T Germer
- Department of Surgery I, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Martin Gasser
- Department of Surgery I, University of Wuerzburg, 97080 Wuerzburg, Germany.
| | - Ana Maria Waaga-Gasser
- Department of Surgery I, Molecular Oncology and Immunology, University of Wuerzburg, 97080 Wuerzburg, Germany.
- Brigham and Women's Hospital, Transplant Research Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
37
|
Alvarado AG, Lathia JD. Taking a Toll on Self-Renewal: TLR-Mediated Innate Immune Signaling in Stem Cells. Trends Neurosci 2016; 39:463-471. [PMID: 27155992 DOI: 10.1016/j.tins.2016.04.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 02/07/2023]
Abstract
Innate immunity has evolved as the front-line cellular defense mechanism to acutely sense and decisively respond to microenvironmental alterations. The Toll-like receptor (TLR) family activates signaling pathways in response to stimuli and is well-characterized in both resident and infiltrating immune cells during neural inflammation, injury, and degeneration. Innate immune signaling has also been observed in neural cells during development and disease, including in the stem and progenitor cells that build the brain and are responsible for its homeostasis. Recently, the activation of developmental programs in malignant brain tumors has emerged as a driver for growth via cancer stem cells. In this review we discuss how innate immune signaling interfaces with stem cell maintenance in the normal and neoplastic brain.
Collapse
Affiliation(s)
- Alvaro G Alvarado
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
38
|
Fehri E, Ennaifer E, Bel Haj Rhouma R, Guizani-Tabbane L, Guizani I, Boubaker S. The role of Toll-like receptor 9 in gynecologic cancer. Curr Res Transl Med 2016; 64:155-159. [PMID: 27765276 DOI: 10.1016/j.retram.2016.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/27/2016] [Indexed: 12/14/2022]
Abstract
Toll-like receptor 9 (TLR9) plays a major role in the fight against DNA viruses infections. Despite its antitumor properties, inappropriate activation of TLR9 during chronic inflammation may cause the activation of transcription factors inducing pro-cancerous activities. Thus, the relationship between TLR9 and cancer remains highly confrontational especially in gynecological cancers and cervical cancer induced by viruses. In this review, we focus on the beneficial and detrimental role of TLR9 in gynecological carcinogenesis. TLR9 contributes to tumor regression by inducing cytotoxic T cell response (CTL), reducing the numbers of myeloid-derived suppressor cells (MDSCs), the tumor-associated macrophages (TAMs) and the regulatory T cells (T regs). It can however, also promote tumor progression and invasiveness of cervical tissue. Therefore, the dichotomous role of TLR9 needs to be carefully investigated in the setting of neoplastic disease.
Collapse
Affiliation(s)
- E Fehri
- HPV Unit Research, Laboratory of Molecular Epidemiology and Experimental Pathology Applied to Infectious Diseases, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia; Department of Human and Experimental Pathology, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia.
| | - E Ennaifer
- HPV Unit Research, Laboratory of Molecular Epidemiology and Experimental Pathology Applied to Infectious Diseases, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia; Department of Human and Experimental Pathology, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia
| | - R Bel Haj Rhouma
- HPV Unit Research, Laboratory of Molecular Epidemiology and Experimental Pathology Applied to Infectious Diseases, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia
| | - L Guizani-Tabbane
- Laboratory of Medical Parasitology Biotechnology and Biomolecules, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia
| | - I Guizani
- HPV Unit Research, Laboratory of Molecular Epidemiology and Experimental Pathology Applied to Infectious Diseases, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia
| | - S Boubaker
- Department of Human and Experimental Pathology, Pasteur Institute of Tunis, BP 74, 1002, le Belvédère, Tunis, Tunisia
| |
Collapse
|
39
|
Schnurr M, Duewell P, Bauer C, Rothenfusser S, Lauber K, Endres S, Kobold S. Strategies to relieve immunosuppression in pancreatic cancer. Immunotherapy 2016; 7:363-76. [PMID: 25917628 DOI: 10.2217/imt.15.9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Despite continuous progress in the understanding of deregulated pathways in pancreatic cancer cells and development of targeted therapies, therapeutic advances with clinical benefit have been scarce over the last decades. The recent success of immunotherapy for some solid cancers has fueled optimism that this approach might also work for pancreatic cancer. However, a highly immunosuppressive microenvironment mediated by tumor, stromal and immune cells creates a major hurdle for immunotherapy. Mouse models have helped to unravel critical immunosuppressive mechanisms that could serve as novel therapeutic targets. Here we review new promising strategies that alone or in combination with other modalities, such as chemotherapy or irradiation, have the potential to lead to tumor immune control and finally better clinical outcome.
Collapse
Affiliation(s)
- Max Schnurr
- Division of Clinical Pharmacology & Center for Integrated Protein Science Munich (CIPSM), Klinikum der Universität München, Munich, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Greco SH, Tomkötter L, Vahle AK, Rokosh R, Avanzi A, Mahmood SK, Deutsch M, Alothman S, Alqunaibit D, Ochi A, Zambirinis C, Mohaimin T, Rendon M, Levie E, Pansari M, Torres-Hernandez A, Daley D, Barilla R, Pachter HL, Tippens D, Malik H, Boutajangout A, Wisniewski T, Miller G. TGF-β Blockade Reduces Mortality and Metabolic Changes in a Validated Murine Model of Pancreatic Cancer Cachexia. PLoS One 2015; 10:e0132786. [PMID: 26172047 PMCID: PMC4501823 DOI: 10.1371/journal.pone.0132786] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 06/18/2015] [Indexed: 01/05/2023] Open
Abstract
Cancer cachexia is a debilitating condition characterized by a combination of anorexia, muscle wasting, weight loss, and malnutrition. This condition affects an overwhelming majority of patients with pancreatic cancer and is a primary cause of cancer-related death. However, few, if any, effective therapies exist for both treatment and prevention of this syndrome. In order to develop novel therapeutic strategies for pancreatic cancer cachexia, appropriate animal models are necessary. In this study, we developed and validated a syngeneic, metastatic, murine model of pancreatic cancer cachexia. Using our model, we investigated the ability of transforming growth factor beta (TGF-β) blockade to mitigate the metabolic changes associated with cachexia. We found that TGF-β inhibition using the anti-TGF-β antibody 1D11.16.8 significantly improved overall mortality, weight loss, fat mass, lean body mass, bone mineral density, and skeletal muscle proteolysis in mice harboring advanced pancreatic cancer. Other immunotherapeutic strategies we employed were not effective. Collectively, we validated a simplified but useful model of pancreatic cancer cachexia to investigate immunologic treatment strategies. In addition, we showed that TGF-β inhibition can decrease the metabolic changes associated with cancer cachexia and improve overall survival.
Collapse
Affiliation(s)
- Stephanie H. Greco
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Lena Tomkötter
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Anne-Kristin Vahle
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Rae Rokosh
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Antonina Avanzi
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Syed Kashif Mahmood
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Michael Deutsch
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Sara Alothman
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Dalia Alqunaibit
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Atsuo Ochi
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Constantinos Zambirinis
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Tasnima Mohaimin
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Mauricio Rendon
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Elliot Levie
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Mridul Pansari
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Alejandro Torres-Hernandez
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Donnele Daley
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Rocky Barilla
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - H. Leon Pachter
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Daniel Tippens
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Hassan Malik
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
| | - Allal Boutajangout
- Department of Neurology, New York University School of Medicine, New York, New York, United States of America
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, New York, New York, United States of America
| | - George Miller
- Department of Surgery, New York University School of Medicine, New York, New York, United States of America
- Department of Cell Biology, New York University School of Medicine, New York, New York, United States of America
| |
Collapse
|
41
|
Gene expression disorders of innate antibacterial signaling pathway in pancreatic cancer patients: implications for leukocyte dysfunction and tumor progression. Cent Eur J Immunol 2014; 39:498-507. [PMID: 26155170 PMCID: PMC4439963 DOI: 10.5114/ceji.2014.47736] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 09/30/2014] [Indexed: 01/01/2023] Open
Abstract
The study was carried out to investigate changes in gene expression of innate antibacterial signaling pathways in patients with pancreatic cancer. Expression of the following genes was measured in peripheral blood leukocytes of 55 patients with pancreatic adenocarcinoma using real-time polymerase chain reaction (RT-PCR): TLR4, NOD1, MyD88, TRAF6 and HMGB1. The levels of expression of TLR4, NOD1 and TRAF6 genes were significantly elevated (p = 0.007; p = 0.001 and p = 0.01, respectively), while MyD88 expression was markedly reduced (p = 0.0002), as compared to controls. Expression of TLR4 and NOD1 exceeded the normal level more than 3.5-fold and there was a significant correlation found between the expression of these genes (r = 0.558, p < 0.001). TLR4, NOD1 and MyD88 genes were expressed at a similar level both before and after surgery. No significant changes in the expression of HMGB1 gene were observed. The results of the study clearly indicate abnormal expression of genes belonging to innate antibacterial signaling pathways in peripheral blood leukocytes of patients with pancreatic cancer, which may lead to leukocyte dysfunction. Overexpression of TLR4, NOD1 and TRAF6 genes, and decreased MyD88 gene expression may contribute to chronic inflammation and tumor progression by up-regulation of the innate antibacterial response. The parameters tested are useful for monitoring innate immunity gene disorders and pancreatic cancer progression.
Collapse
|
42
|
Cheng S, Liu J, Zhang Y, Lin Y, Liu Q, Li H, Huang J, Zhang P. Association detection between genetic variants in the microRNA binding sites of toll-like receptors signaling pathway genes and bladder cancer susceptibility. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8118-8126. [PMID: 25550860 PMCID: PMC4270586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 10/18/2014] [Indexed: 06/04/2023]
Abstract
Bladder cancer (BCa) is the second most common urological malignancy, and the incidence of BCa has dramatically increased recently. Various toll-like receptors (TLRs) signaling pathway proteins were proven to be associated with BCa susceptibility. However, the effect of genetic variants in TLRs signaling pathway genes on risk of BCa has not been elucidated clearly. Previous studies mainly focused on the coding region of target genes, while in this study, polymorphisms in the non-coding region, microRNA (miRNA) binding sites were investigated as potential targets. We used bioinformatics approach to screen 100 BCa related TLRs signaling pathway genes. Candidate polymorphisms were select in this region and 8 polymorphisms were confirmed. Rs72552316, located at the 3'UTR of the TLR7 gene, exhibited significant association with risk of BCa, indicating a strong relationship with decreased risk of BCa (P ≤ 0.0001). Furthermore, no association was detected between all the polymorphisms and recurrence-free survival time of overall study population or non-muscle invasive BCa subgroups. In conclusion, rs72552316 in the miRNA binding sites of TLR7 might contribute to BCa susceptibility, and this finding provided new targets for high BCa risk population screening.
Collapse
Affiliation(s)
- Sihang Cheng
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Jiaming Liu
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Yonggang Zhang
- The Periodical Press of West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Yifei Lin
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Qinyu Liu
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Hong Li
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Jin Huang
- West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| | - Peng Zhang
- Department of Urology, West China Hospital, Sichuan UniversityGuoxuexiang 37, Chengdu 610041, Sichuan, China
| |
Collapse
|
43
|
Purine-metabolizing ectoenzymes control IL-8 production in human colon HT-29 cells. Mediators Inflamm 2014; 2014:879895. [PMID: 25242873 PMCID: PMC4158428 DOI: 10.1155/2014/879895] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 07/03/2014] [Indexed: 12/20/2022] Open
Abstract
Interleukin-8 (IL-8) plays key roles in both chronic inflammatory diseases and tumor modulation. We previously observed that IL-8 secretion and function can be modulated by nucleotide (P2) receptors. Here we investigated whether IL-8 release by intestinal epithelial HT-29 cells, a cancer cell line, is modulated by extracellular nucleotide metabolism. We first identified that HT-29 cells regulated adenosine and adenine nucleotide concentration at their surface by the expression of the ectoenzymes NTPDase2, ecto-5′-nucleotidase, and adenylate kinase. The expression of the ectoenzymes was evaluated by RT-PCR, qPCR, and immunoblotting, and their activity was analyzed by RP-HPLC of the products and by detection of Pi produced from the hydrolysis of ATP, ADP, and AMP. In response to poly (I:C), with or without ATP and/or ADP, HT-29 cells released IL-8 and this secretion was modulated by the presence of NTPDase2 and adenylate kinase. Taken together, these results demonstrate the presence of 3 ectoenzymes at the surface of HT-29 cells that control nucleotide levels and adenosine production (NTPDase2, ecto-5′-nucleotidase and adenylate kinase) and that P2 receptor-mediated signaling controls IL-8 release in HT-29 cells which is modulated by the presence of NTPDase2 and adenylate kinase.
Collapse
|