1
|
Hsu CY, Jasim SA, Rasool KH, H M, Kaur J, Jabir MS, Alhajlah S, Kumar A, Jawad SF, Husseen B. Divergent functions of TLRs in gastrointestinal (GI) cancer: Overview of their diagnostic, prognostic and therapeutic value. Semin Oncol 2025; 52:152344. [PMID: 40347779 DOI: 10.1016/j.seminoncol.2025.152344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 05/14/2025]
Abstract
The relationship between the innate immune signal and the start of the adaptive immune response is the central idea of this theory. By controlling the inflammatory and tissue-repair reactions to damage, the Toll-like receptors (TLRs), as a family of PRRs, have attracted increasing attention for its function in protecting the host against infection and preserving tissue homeostasis. Microbial infection, damage, inflammation, and tissue healing have all been linked to the development of malignancies, especially gastrointestinal (GI) cancers. Recently, increased studies on TLR recognition and binding, as well as their ligands, have significantly advanced our knowledge of the various TLR signaling pathways and offered therapy options for GI malignancies. Upon activation by pathogen-associated or damage-associated molecular patterns (DAMPs and PAMPs), TLRs trigger key pathways like NF-κB, MAPK, and IRF. NF-κB activation promotes inflammation, cell survival, and proliferation, often contributing to tumor growth, metastasis, and therapy resistance. MAPK pathways similarly drive uncontrolled cell growth and invasion, while IRF pathways modulate interferon production, yielding both anti-tumor and protumor effects. The resulting chronic inflammatory environment within tumors can foster progression, yet TLR activation can also stimulate beneficial anti-tumor immune responses. However, the functions of TLR expression in GI cancers and their diagnostic and prognostic along with therapeutic value have not yet entirely been elucidated. Understanding how TLR activation contributes to anti-cancer immunity against GI malignancies may hasten immunotherapy developments and increase patient survival.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, USA
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Anbar, Iraq; Biotechnology Department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | - Khetam Habeeb Rasool
- Department of Biology, College of Science, University of Mustansiriyah, Mustansiriyah, Iraq
| | - Malathi H
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Jaswinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Mohali, Punjab, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Anbar, Iraq
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra, Saudi Arabia.
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after the First President of Russia Boris Yeltsin, Ekaterinburg, Russia; Centre for Research Impact & Outcome, Chitkara University, Rajpura, Punjab, India; Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Sabrean F Jawad
- Department of Pharmacy, Al-Mustaqbal University College, Hillah, Babylon, Iraq
| | - Beneen Husseen
- Medical Laboratory Technique College, the Islamic University, Najaf, Iraq; Medical Laboratory Technique College, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Medical Laboratory Technique College, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
2
|
Cheng PTM, Topham JT, Aldeheshi A, Taylor GA, Pleasance E, McConechy MK, Nelson JMT, Schaeffer DF, Jones SJM, Marra MA, Laskin J, Renouf DJ. Whole-genome analysis of an aggressive metastatic pancreatic solid pseudopapillary neoplasm. NPJ Precis Oncol 2025; 9:59. [PMID: 40038436 PMCID: PMC11880304 DOI: 10.1038/s41698-025-00843-7] [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: 11/13/2024] [Accepted: 02/20/2025] [Indexed: 03/06/2025] Open
Abstract
Pancreatic solid pseudopapillary neoplasms (SPNs) are uncommon tumors that rarely exhibit aggressive behavior. Given disease rarity, comprehensive studies to understand tumor biology, clinical course, and optimal management are limited. We describe an unusual case of a 55-year-old man with metastatic pancreatic SPN, where whole-genome and transcriptome analyses of the primary tumor and a metastatic liver lesion revealed a shared homozygous non-canonical mutation in APC. The patient received upfront modified FOLFIRINOX (infusional 5-fluorouracil, irinotecan, and oxaliplatin) chemotherapy due to rapidly progressive symptoms, demonstrating an early and sustained treatment response. Therefore, we identified potential genetic determinants of tumorigenesis and progression in a pathologically and clinically aggressive SPN, which may have important prognostic and treatment implications.
Collapse
Affiliation(s)
- Phoebe T M Cheng
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada.
| | - James T Topham
- Pancreas Centre BC, Vancouver, BC, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Ayman Aldeheshi
- Department, Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gregory A Taylor
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Melissa K McConechy
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Jessica M T Nelson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - David F Schaeffer
- Pancreas Centre BC, Vancouver, BC, Canada
- Department, Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Daniel J Renouf
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada.
- Pancreas Centre BC, Vancouver, BC, Canada.
| |
Collapse
|
3
|
Yang Y, Jin C, Yeo A, Jin B. Multiple Factors Determine the Oncolytic or Carcinogenic Effects of TLRs Activation in Cancer. J Immunol Res 2024; 2024. [DOI: 10.1155/2024/1111551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 12/13/2023] [Indexed: 01/05/2025] Open
Abstract
Toll‐like receptors (TLRs) belong to a germline‐encoded protein family. These are pattern recognition receptors. They sense pathogen‐associated molecular patterns (PAMPs). When this occurs, activation of the NF‐ĸB pathway follows. This triggers the innate immune response of the host. The consequent inflammatory cytokine response usually contributes to the elimination of the pathogen. Activation of TLRs also induces an adaptive immune response by a cross‐prime mechanism. This mechanism is employed in cancer immunotherapy. Using TLR ligands as adjuvants induces upregulation of costimulatory signals which in turn activates a cytotoxic leukocyte response against cancer cells. However, TLRs are also overexpressed in human cancer cells resulting in increased cell proliferation, migration, invasion, and angiogenesis. An intracellular adaptor, myeloid differentiation factor 88 (MyD88) probably mediates this process. MyD88 is intimately involved with all TLRs except TLR3. One consequence of the interaction between a TLR and MyD88 is activation of NF‐ĸB. In this context of a variety of proinflammtory cytokines being produced, chronic inflammation may result. Inflammation is an important protective mechanism. However, chronic inflammation is also involved in carcinogenesis. Activation of NF‐ĸB inhibits apoptosis and under certain circumstances, tumor cell survival. In this review, the potential therapeutic value of TLRs in immunotherapy and its role in oncogenesis are explored. The emerging use of artificial intelligence is mentioned.
Collapse
|
4
|
Huang KCY, Ke TW, Chen JY, Hong WZ, Chiang SF, Lai CY, Chen TW, Yang PC, Chen LC, Liang JA, Chen WTL, Chao KSC. Dysfunctional TLR1 reduces the therapeutic efficacy of chemotherapy by attenuating HMGB1-mediated antitumor immunity in locally advanced colorectal cancer. Sci Rep 2023; 13:19440. [PMID: 37945630 PMCID: PMC10636035 DOI: 10.1038/s41598-023-46254-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/30/2023] [Indexed: 11/12/2023] Open
Abstract
Regional lymph node metastasis is an important predictor for survival outcome and an indicator for postoperative adjuvant chemotherapy in patients with colorectal cancer. Even with advances in adjuvant chemotherapeutic regimens, 5-year distant metastasis and survival rates are still unsatisfactory. Here, we evaluate the clinical significance of polymorphisms in receptors for HMGB1, which is the hallmark of chemotherapy-induced immunogenic cell death, in patients with stage II-III colon carcinoma (COAD). We found that high cytosolic HMGB1 is elicited in stage III COAD patients who received adjuvant chemotherapy. Patients with the TLR1-N248S polymorphism (rs4833095), which causes loss-of-function in HMGB1-mediated TLR1-TLR2 signaling, may influence the therapeutic efficacy of adjuvant chemotherapy, leading to a high risk of distant metastasis within 5 years [HR = 1.694, 95% CI = 1.063-2.698, p = 0.027], suggesting that TLR1-N248S is an independent prognostic factor for locally advanced colon carcinoma patients. We found that defective TLR1 impaired TLR1/2 signaling during dendritic cell (DC) maturation for the antitumor immune response under immunogenic chemotherapy oxaliplatin (OXP) treatment. Defective TLR1 on DCs impaired their maturation ability by HMGB1 and reduced the secretion of IFNγ from T cells to eradicate tumor cells in vitro. Moreover, systemic inhibition of TLR1/2 dramatically reduced the tumor-infiltrating immune cells by OXP treatment, leading to poor therapeutic response to OXP. In contrast, administration of a TLR1/2 agonist synergistically increased the benefit of OXP treatment and triggered a high density of tumor-infiltrating immune cells. We also observed that fewer tumor-infiltrating cytotoxic T lymphocytes were located within the tumor microenvironment in patients bearing the TLR1-N248S polymorphism. Overall, our results suggest that dysfunctional TLR1 may reduce the therapeutic response to adjuvant chemotherapy by impairing HMGB1-mediated DC maturation and attenuating the antitumor immune response in locally advanced colon carcinoma patients.
Collapse
Affiliation(s)
- Kevin Chih-Yang Huang
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Tao-Wei Ke
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- School of Chinese Medicine and Graduate Institute of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Jia-Yi Chen
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Wei-Ze Hong
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Shu-Fen Chiang
- Lab of Precision Medicine, Feng-Yuan Hospital, Ministry of Health and Welfare, Taichung, 42055, Taiwan, ROC
| | - Chia-Ying Lai
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, 40402, Taiwan, ROC
- Translation Research Core, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Tsung-Wei Chen
- Department of Pathology, Asia University Hospital, Asia University, Taichung, 41354, Taiwan, ROC
| | - Pei-Chen Yang
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Liang-Chi Chen
- Department of Pathology, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC
| | - Ji-An Liang
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC
- Department of Radiotherapy, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC
| | - William Tzu-Liang Chen
- Department of Colorectal Surgery, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC.
- Department of Colorectal Surgery, China Medical University HsinChu Hospital, China Medical University, HsinChu, 302, Taiwan, ROC.
- Department of Surgery, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC.
| | - K S Clifford Chao
- Proton Therapy and Science Center, China Medical University Hospital, China Medical University, Taichung, 40402, Taiwan, ROC.
- Department of Radiation Oncology, China Medical University Hospital, China Medical University, Taichung, Taiwan, ROC.
- Department of Radiotherapy, School of Medicine, China Medical University, Taichung, 40402, Taiwan, ROC.
| |
Collapse
|
5
|
Sun H, Li Y, Zhang P, Xing H, Zhao S, Song Y, Wan D, Yu J. Targeting toll-like receptor 7/8 for immunotherapy: recent advances and prospectives. Biomark Res 2022; 10:89. [PMID: 36476317 PMCID: PMC9727882 DOI: 10.1186/s40364-022-00436-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022] Open
Abstract
Toll-like receptors (TLRs) are a large family of proteins that are expressed in immune cells and various tumor cells. TLR7/8 are located in the intracellular endosomes, participate in tumor immune surveillance and play different roles in tumor growth. Activation of TLRs 7 and 8 triggers induction of a Th1 type innate immune response in the highly sophisticated process of innate immunity signaling with the recent research advances involving the small molecule activation of TLR 7 and 8. The wide range of expression and clinical significance of TLR7/TLR8 in different kinds of cancers have been extensively explored. TLR7/TLR8 can be used as novel diagnostic biomarkers, progression and prognostic indicators, and immunotherapeutic targets for various tumors. Although the mechanism of action of TLR7/8 in cancer immunotherapy is still incomplete, TLRs on T cells are involved in the regulation of T cell function and serve as co-stimulatory molecules and activate T cell immunity. TLR agonists can activate T cell-mediated antitumor responses with both innate and adaptive immune responses to improve tumor therapy. Recently, novel drugs of TLR7 or TLR8 agonists with different scaffolds have been developed. These agonists lead to the induction of certain cytokines and chemokines that can be applied to the treatment of some diseases and can be used as good adjutants for vaccines. Furthermore, TLR7/8 agonists as potential therapeutics for tumor-targeted immunotherapy have been developed. In this review, we summarize the recent advances in the development of immunotherapy strategies targeting TLR7/8 in patients with various cancers and chronic hepatitis B.
Collapse
Affiliation(s)
- Hao Sun
- Department of Radiotherapy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yingmei Li
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Peng Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Haizhou Xing
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yongping Song
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Dingming Wan
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004 Henan China
| |
Collapse
|
6
|
Li L, Lu S, Ma C. Anti-proliferative and pro-apoptotic effects of curcumin on skin cutaneous melanoma: Bioinformatics analysis and in vitro experimental studies. Front Genet 2022; 13:983943. [PMID: 36171883 PMCID: PMC9510772 DOI: 10.3389/fgene.2022.983943] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022] Open
Abstract
Objective: To reveal the potential mechanisms of curcumin for the treatment of skin cutaneous melanoma (SKCM) and its identify novel prognostic biomarkers. Methods: We searched the Cancer Genome Atlas and Traditional Chinese Medicine Systems Pharmacology database for the data on SKCM and curcumin. We conducted data analysis using R and online tools. The propagation and migration of SKCM cells were assessed with CCK-8 and scratch wound assays, respectively. We assessed apoptosis by TUNEL assay and western blot. Results: The survival analysis revealed that the mRNA expressions of DPYD, DPYS, LYN, PRKCQ, and TLR1 were significantly related to a favorable overall survival in SKCM patients. Additionally, the mRNA expression level of DPYD was associated with GPI, LYN, PCSK9, PRKCQ, and TLR1 mRNAs. GSEA results showed that the prognostic hub genes were augmented with ultraviolet, apoptosis, and metastasis. Curcumin expressed proliferation and migration of SK-MEL-1 cells (p < 0.05), and induced apoptosis (p < 0.05) significantly. Conclusion: Curcumin may have potential therapeutic effects in SKCM by inhibiting cell proliferation and migration and inducing apoptosis by regulating oxygen-related signaling pathways. The hub genes might be identified as novel biomarkers for SKCM.
Collapse
Affiliation(s)
- Long Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuwen Lu
- Department of Ophthalmology, the First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Chao Ma
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Chao Ma,
| |
Collapse
|
7
|
Ye J, Mills BN, Qin SS, Garrett-Larsen J, Murphy JD, Uccello TP, Han BJ, Vrooman TG, Johnston CJ, Lord EM, Belt BA, Linehan DC, Gerber SA. Toll-like receptor 7/8 agonist R848 alters the immune tumor microenvironment and enhances SBRT-induced antitumor efficacy in murine models of pancreatic cancer. J Immunother Cancer 2022; 10:e004784. [PMID: 35851308 PMCID: PMC9295644 DOI: 10.1136/jitc-2022-004784] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Stereotactic body radiotherapy (SBRT) has been increasingly used as adjuvant therapy in pancreatic ductal adenocarcinoma (PDAC), and induces immunogenic cell death, which leads to the release of tumor antigen and damage-associated molecular patterns. However, this induction often fails to generate sufficient response to overcome pre-existing tumor microenvironment (TME) immunosuppression. Toll-like receptor (TLR) 7/8 ligands, such as R848, can amplify the effect of tumor vaccines, with recent evidence showing its antitumor effect in pancreatic cancer by modulating the immunosuppressive TME. Therefore, we hypothesized that the combination of R848 and SBRT would improve local and systemic antitumor immune responses by potentiating the antitumor effects of SBRT and reversing the immunosuppressive nature of the PDAC TME. METHODS Using murine models of orthotopic PDAC, we assessed the combination of intravenous TLR7/8 agonist R848 and local SBRT on tumor growth and immune response in primary pancreatic tumors. Additionally, we employed a hepatic metastatic model to investigate if the combination of SBRT targeting only the primary pancreatic tumor and systemic R848 is effective in controlling established liver metastases. RESULTS We demonstrated that intravenous administration of the TLR7/8 agonist R848, in combination with local SBRT, leads to superior tumor control compared with either treatment alone. The combination of R848 and SBRT results in significant immune activation of the pancreatic TME, including increased tumor antigen-specific CD8+ T cells, decreased regulatory T cells, and enhanced antigen-presenting cells maturation, as well as increased interferon gamma, granzyme B, and CCL5 along with decreased levels of interleukin 4 (IL-4), IL-6, and IL-10. Importantly, the combination of SBRT and systemic R848 also resulted in similar immunostimulatory changes in liver metastases, leading to improved metastatic control. CD8+ T cell depletion studies highlighted the necessity of these effector cells at both the local and hepatic metastatic sites. T cell receptor (TCR) clonotype analysis indicated that systemic R848 not only diversified the TCR repertoire but also conditioned the metastatic foci to facilitate entry of CD8+ T cells generated by SBRT therapy. CONCLUSIONS These findings suggest that systemic administration of TLR7/8 agonists in combination with SBRT may be a promising avenue for metastatic PDAC treatment.
Collapse
Affiliation(s)
- Jian Ye
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Bradley N Mills
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Shuyang S Qin
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jesse Garrett-Larsen
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - Joseph D Murphy
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Taylor P Uccello
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Booyeon J Han
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Tara G Vrooman
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Carl J Johnston
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York, USA
| | - Edith M Lord
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Brian A Belt
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
| | - David C Linehan
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | - Scott A Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, New York, USA
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, New York, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
8
|
Barfield R, Qu C, Steinfelder RS, Zeng C, Harrison TA, Brezina S, Buchanan DD, Campbell PT, Casey G, Gallinger S, Giannakis M, Gruber SB, Gsur A, Hsu L, Huyghe JR, Moreno V, Newcomb PA, Ogino S, Phipps AI, Slattery ML, Thibodeau SN, Trinh QM, Toland AE, Hudson TJ, Sun W, Zaidi SH, Peters U. Association between germline variants and somatic mutations in colorectal cancer. Sci Rep 2022; 12:10207. [PMID: 35715570 PMCID: PMC9205954 DOI: 10.1038/s41598-022-14408-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 06/07/2022] [Indexed: 01/11/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease with evidence of distinct tumor types that develop through different somatically altered pathways. To better understand the impact of the host genome on somatically mutated genes and pathways, we assessed associations of germline variations with somatic events via two complementary approaches. We first analyzed the association between individual germline genetic variants and the presence of non-silent somatic mutations in genes in 1375 CRC cases with genome-wide SNPs data and a tumor sequencing panel targeting 205 genes. In the second analysis, we tested if germline variants located within previously identified regions of somatic allelic imbalance were associated with overall CRC risk using summary statistics from a recent large scale GWAS (n≃125 k CRC cases and controls). The first analysis revealed that a variant (rs78963230) located within a CNA region associated with TLR3 was also associated with a non-silent mutation within gene FBXW7. In the secondary analysis, the variant rs2302274 located in CDX1/PDGFRB frequently gained/lost in colorectal tumors was associated with overall CRC risk (OR = 0.96, p = 7.50e-7). In summary, we demonstrate that an integrative analysis of somatic and germline variation can lead to new insights about CRC.
Collapse
Affiliation(s)
- Richard Barfield
- grid.26009.3d0000 0004 1936 7961Department of Biostatistics and Bioinformatics, Duke University, 11028A Hock Plaza, 2424 Erwin Road Suite 1106, Durham, NC 27705 USA
| | - Conghui Qu
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Robert S. Steinfelder
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Chenjie Zeng
- grid.280128.10000 0001 2233 9230National Human Genome Research Institute, National Institutes of Health, Bethesda, MD USA
| | - Tabitha A. Harrison
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Stefanie Brezina
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - Daniel D. Buchanan
- grid.1008.90000 0001 2179 088XColorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1008.90000 0001 2179 088XUniversity of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, VIC 3010 Australia ,grid.416153.40000 0004 0624 1200Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, VIC Australia
| | - Peter T. Campbell
- grid.251993.50000000121791997Department of Epidemiology and Population Science, Albert Einstein College of Medicine, Bronx, NY USA
| | - Graham Casey
- grid.27755.320000 0000 9136 933XCenter for Public Health Genomics, University of Virginia, Charlottesville, VA USA
| | - Steven Gallinger
- grid.250674.20000 0004 0626 6184Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, ON Canada
| | - Marios Giannakis
- grid.65499.370000 0001 2106 9910Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA USA ,grid.66859.340000 0004 0546 1623The Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Stephen B. Gruber
- grid.42505.360000 0001 2156 6853Department of Medical Oncology and Therapeuytic, University of Southern California, Los Angeles, CA USA
| | - Andrea Gsur
- grid.22937.3d0000 0000 9259 8492Institute of Cancer Research, Department of Medicine I, Medical University Vienna, Vienna, Austria
| | - Li Hsu
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ,grid.34477.330000000122986657Department of Biostatistics, University of Washington, Seattle, WA USA
| | - Jeroen R. Huyghe
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA
| | - Victor Moreno
- grid.418701.b0000 0001 2097 8389Oncology Data Analytics Program, Catalan Institute of Oncology-IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain ,grid.466571.70000 0004 1756 6246CIBER Epidemiología Y Salud Pública (CIBERESP), Madrid, Spain ,grid.5841.80000 0004 1937 0247Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain ,grid.418284.30000 0004 0427 2257ONCOBEL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Polly A. Newcomb
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ,grid.34477.330000000122986657School of Public Health, University of Washington, Seattle, WA USA
| | - Shuji Ogino
- grid.66859.340000 0004 0546 1623The Broad Institute of MIT and Harvard, Cambridge, MA USA ,grid.38142.3c000000041936754XProgram in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA ,Cancer Immunology Program, Dana-Farber Harvard Cancer Center, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Amanda I. Phipps
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ,Department of Epidemiology, Fred Hutchinson Cancer Research Center, University of Washington, 1100 Fairview Ave N, Mail Stop M4-B402, Seattle, WA 98109 USA
| | - Martha L. Slattery
- grid.223827.e0000 0001 2193 0096Department of Internal Medicine, University of Utah, Salt Lake City, UT USA
| | - Stephen N. Thibodeau
- grid.66875.3a0000 0004 0459 167XDivision of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN USA
| | - Quang M. Trinh
- grid.419890.d0000 0004 0626 690XOntario Institute for Cancer Research, Toronto, ON Canada
| | - Amanda E. Toland
- grid.261331.40000 0001 2285 7943Departments of Cancer Biology and Genetics and Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, OH USA
| | - Thomas J. Hudson
- grid.419890.d0000 0004 0626 690XOntario Institute for Cancer Research, Toronto, ON Canada
| | - Wei Sun
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ,grid.34477.330000000122986657Department of Biostatistics, University of Washington, Seattle, WA USA ,grid.410711.20000 0001 1034 1720Department of Biostatistics, University of North Carolina, Chapel Hill, NC USA
| | - Syed H. Zaidi
- grid.419890.d0000 0004 0626 690XOntario Institute for Cancer Research, Toronto, ON Canada
| | - Ulrike Peters
- grid.270240.30000 0001 2180 1622Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA USA ,Department of Epidemiology, Fred Hutchinson Cancer Research Center, University of Washington, 1100 Fairview Ave N, Mail Stop M4-B402, Seattle, WA 98109 USA
| |
Collapse
|
9
|
Exploration of the System-Level Mechanisms of the Herbal Drug FDY003 for Pancreatic Cancer Treatment: A Network Pharmacological Investigation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7160209. [PMID: 35591866 PMCID: PMC9113891 DOI: 10.1155/2022/7160209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/12/2022] [Indexed: 11/18/2022]
Abstract
Pancreatic cancer (PC) is the most lethal cancer with the lowest survival rate globally. Although the prescription of herbal drugs against PC is gaining increasing attention, their polypharmacological therapeutic mechanisms are yet to be fully understood. Based on network pharmacology, we explored the anti-PC properties and system-level mechanisms of the herbal drug FDY003. FDY003 decreased the viability of human PC cells and strengthened their chemosensitivity. Network pharmacological analysis of FDY003 indicated the presence of 16 active phytochemical components and 123 PC-related pharmacological targets. Functional enrichment analysis revealed that the PC-related targets of FDY003 participate in the regulation of cell growth and proliferation, cell cycle process, cell survival, and cell death. In addition, FDY003 was shown to target diverse key pathways associated with PC pathophysiology, namely, the PIK3-Akt, MAPK, FoxO, focal adhesion, TNF, p53, HIF-1, and Ras pathways. Our network pharmacological findings advance the mechanistic understanding of the anti-PC properties of FDY003 from a system perspective.
Collapse
|
10
|
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
|
11
|
Lian L, Teng SB, Xia YY, Shen XM, Zheng Y, Han SG, Wang WJ, Xu XF, Zhou C. Development and verification of a hypoxia- and immune-associated prognosis signature for esophageal squamous cell carcinoma. J Gastrointest Oncol 2022; 13:462-477. [PMID: 35557566 PMCID: PMC9086047 DOI: 10.21037/jgo-22-69] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/08/2022] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Esophageal cancer is one of the most common gastrointestinal malignancies worldwide, with high morbidity and mortality in China. The clinical importance of the interaction between hypoxia and immune status in the tumor microenvironment has been established in esophageal squamous cell carcinoma (ESCC). This study aims to develop a new hypoxia- and immune-based gene signature to predict the survival of ESCC patients. METHODS The RNA-sequencing and clinical data of 173 cases of ESCC and 271 normal tissues were obtained from The Cancer Genome Atlas (TCGA) data portal and the Genotype-Tissue Expression (GTEx) database. Hypoxia-related genes (HRGs) and immune-related genes (IRGs) were retrieved from publicly shared data. Differentially expressed gene (DEG) analyses were carried out by the DESeq2 method using the edgeR package in R. Based on the intersection of the DEGs and HRGs/IRGs, differentially expressed HRGs (DEHRGs) and differentially expressed IRGs (DEIRGs) were obtained. DEHRGs and DEIRGs associated with prognosis were evaluated using univariate Cox proportional hazards analysis. A prognostic risk score model was constructed according to the genes acquired through Cox regression. Univariate analysis and Cox proportional hazards analysis were used to determine the independent prognostic factors related to prognosis. A nomogram was developed to predict the 1-, 2-, and 3-year overall survival (OS) probability. RESULTS A total of 73 intersecting genes were obtained as DEHRGs and a total of 548 intersecting genes were obtained as DEIRGs. The risk score was established using 8 genes (FABP7, TLR1, SYTL1, APLN, OSM, EGFR, IL17RD, MYH9) acquired from univariate Cox analysis. Based on this 8-gene-based risk score, a risk prognosis classifier was constructed to classify the samples into high- and low-risk groups according to the median risk score. The nomogram model was constructed to predict the OS of ESCC patients. CONCLUSIONS The hypoxia- and immune-based gene signature might serve as a prognostic classifier for clinical decision-making regarding individualized management, follow-up plans, and treatment strategies for ESCC patients.
Collapse
Affiliation(s)
- Lian Lian
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Shi-Bing Teng
- Department of Thoracic Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - You-You Xia
- Department of Radiation Oncology, The Affiliated Lianyungang Hospital of Xuzhou Medical University (The First People’s Hospital of Lianyungang), Lianyungang, China
| | - Xiao-Ming Shen
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Yan Zheng
- Department of Oncology, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Shu-Guang Han
- Department of General Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Wen-Jie Wang
- Department of Radio-Oncology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Xue-Fei Xu
- Department of General Surgery, Suzhou Xiangcheng People’s Hospital, Suzhou, China
| | - Chong Zhou
- Department of Radiation Oncology, Xuzhou Central Hospital, Xuzhou, China
| |
Collapse
|
12
|
Lan Y, He L, Dong X, Tang R, Li W, Wang J, Wang L, Yue B, Price M, Guo T, Fan Z. Comparative transcriptomes of three different skin sites for the Asiatic toad ( Bufo gargarizans). PeerJ 2022; 10:e12993. [PMID: 35223212 PMCID: PMC8877344 DOI: 10.7717/peerj.12993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 02/02/2022] [Indexed: 01/11/2023] Open
Abstract
Toads release toxic dry secretions from glands in their skin. Toxin possesses a wide range of biological effects, but little is known about its specific gene expression pattern and regulatory mechanisms. The Asiatic toad (Bufo gargarizans) is widely used to produce toxin. Here, we explored the gene expression of 30 tissue samples from three different skin sites (parotoid gland, dorsal skin, and abdomen skin) of B. gargarizans. After de novo assembly, 783,130 unigenes with an average length of 489 bp (N50 = 556 bp) were obtained. A total of 9,248 significant differentially expressed genes (DEGs) were detected. There were 8,819 DEGs between the parotoid gland and abdomen skin and 1,299 DEGs between the dorsal skin and abdomen skin, while only 1,283 DEGs were obtained between the parotoid gland and dorsal skin. Through enrichment analysis, it was found that the detected differential gene expressions corresponded to the different functions of different skin sites. Our key findings were the genetic expression of toxin secretion, the protection function of skin, and the related genes such as HSD3B, Cyp2c, and CAT, LGALS9. In conclusion, we provide useful transcript resources to study the gene expression and gene function of B. gargarizans and other amphibians. The detected DEGs between different sites of the skin provided better insights into the genetic mechanisms of toxin secretion and the protection function of skin for amphibians.
Collapse
Affiliation(s)
- Yue Lan
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Lewei He
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Xue Dong
- Department of Ambulatory surgery, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruixiang Tang
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Wanyu Li
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Jiao Wang
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Lei Wang
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China,Sichuan Engineering Research Center for Medicinal Animals, Xichang, Sichuan, China
| | - Bisong Yue
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China,Sichuan Engineering Research Center for Medicinal Animals, Xichang, Sichuan, China
| | - Megan Price
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| | - Tao Guo
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, kChengdu, Sichuan, China
| | - Zhenxin Fan
- Key Laboratory of Bioresources and Eco-Environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Lu L, Hu Y, Wang C, Jiang F, Wu C. Methylation and Expression of the Exercise-Related TLR1 Gene Is Associated With Low Grade Glioma Prognosis and Outcome. Front Mol Biosci 2021; 8:747933. [PMID: 34869584 PMCID: PMC8635206 DOI: 10.3389/fmolb.2021.747933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/30/2021] [Indexed: 01/03/2023] Open
Abstract
Background: Exercise improves function, reduces disability, maintains independence, and improves quality of life for low-grade glioma (LGG) patients. Exercise can also improve the effectiveness of cancer treatment. The goal of this research was to find potential exercise related genes that may be used to predict exercise levels and may be used as a biomarker for cancer outcomes. Methods: The GSE111551 database was thoroughly examined in this research, and the resulting conclusion of exercise-related genes was reached. The protein interaction network (PPI) was used to examine the differentially expressed genes (DEGs). Then the exercise-related gene TLR1 was chosen. The expression, methylation degree, prognosis, and immune relevance of TLR1 were investigated using bioinformatics. In addition, we verified the role of TLR1 in Glioma cell lines. Results: LGG patients with reduced TLR1 expression and hypermethylation had a better overall survival (OS) and progression free survival (PFS), using the TCGA database. Low TLR1 expression and hypermethylation of TLR1 were found to be independent biomarkers for OS using Cox regression. Furthermore, the CGGA database was used to confirm the prognostic function of TLR1 in this cancer. Finally, most methylation sites of TLR1 were strongly correlated with immune infiltration and immune checkpoint. Then, reducing TLR1 expression substantially slowed the cell cycle and decreased LGG cell proliferation, emigration, and infiltration in vitro. Conclusions: Exercise-related gene TLR1 has the potential to be a useful prognostic biomarker, and it is thought to be involved in immune cell infiltration and immunotherapy in LGG.
Collapse
Affiliation(s)
- Lichun Lu
- Department of Neurology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Yifang Hu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Wang
- Department of Neurology, The Affiliated Suzhou Science & Technology Town Hospital of Nanjing Medical University, Suzhou, China
| | - Feng Jiang
- Department of Neonatology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chuyan Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
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
|
15
|
Huang Z, He A, Wang J, Lu H, Xu X, Zhang R, Liao W, Feng Q, Wu L. Toll-like receptor 3 is a potential prognosis marker and associated with immune infiltration in stomach adenocarcinoma. Cancer Biomark 2021; 34:77-93. [PMID: 34657879 DOI: 10.3233/cbm-210354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Toll-like receptors participate in various biological mechanisms, mainly including the immune response and inflammatory response. Nevertheless, the role of TLRs in STAD remains unclear. OBJECTIVE We aimed to explore the expression, prognosis performance of TLRs in STAD and their relationship with immune infiltration. METHODS Student's t-test was used to evaluate the expression of TLRs between STAD tissues and normal tissues. Kaplan-Meier method was applied to explored the prognosis value of TLRs in STAD. And qRT-PCR validated their expression and prognosis value. Spearman's correlation analysis and Wilcoxon rank-sum test were used to assess the association between TLRs and immune infiltration in STAD. RESULTS The mRNA level of TLR3 was downregulated in STAD. We summarized genetic mutations and CNV alteration of TLRs in STAD cohort. Prognosis analysis revealed that STAD patients with high TLR3 expression showed better prognosis in OS, FP and PPS. The result of qRT-PCR suggested that TLR3 expression was decreased in STAD tissues and STAD patients with high TLR3 mRNA level had a better OS. Univariate and multivariate cox regression analysis suggested TLR3 expression and clinical stage as independent factors affecting STAD patients' prognosis. A positive association existed between TLR3 expression and the abundance of immune cells and the expression of various immune biomarkers. Furthermore, key targets related to TLR3 were identified in STAD, mainly including MIR-129 (GCAAAAA), PLK1, and V$IRF1_01. CONCLUSIONS Our result demonstrated TLR3 as a prognosis marker and associated with immune infiltration in STAD.
Collapse
Affiliation(s)
- Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Aoxiao He
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.,Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hongcheng Lu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaoyun Xu
- Department of General Surgery, Jinxian People's Hospital, Nanchang, Jiangxi, China
| | - Rongguiyi Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wenjun Liao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qian Feng
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Linquan Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
16
|
Wang X, Chen Z, Murani E, D'Alessandro E, An Y, Chen C, Li K, Galeano G, Wimmers K, Song C. A 192 bp ERV fragment insertion in the first intron of porcine TLR6 may act as an enhancer associated with the increased expressions of TLR6 and TLR1. Mob DNA 2021; 12:20. [PMID: 34407874 PMCID: PMC8375133 DOI: 10.1186/s13100-021-00248-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 07/23/2021] [Indexed: 12/20/2022] Open
Abstract
Background Toll-like receptors (TLRs) play important roles in building innate immune and inducing adaptive immune responses. Associations of the TLR genes polymorphisms with disease susceptibility, which are the basis of molecular breeding for disease resistant animals, have been reported extensively. Retrotransposon insertion polymorphisms (RIPs), as a new type of molecular markers developed recently, have great potential in population genetics and quantitative trait locus mapping. In this study, bioinformatic prediction combined with PCR-based amplification was employed to screen for RIPs in porcine TLR genes. Their population distribution was examined, and for one RIP the impact on gene activity and phenotype was further evaluated. Results Five RIPs, located at the 3' flank of TLR3, 5' flank of TLR5, intron 1 of TLR6, intron 1 of TLR7, and 3' flank of TLR8 respectively, were identified. These RIPs were detected in different breeds with an uneven distribution among them. By using the dual luciferase activity assay a 192 bp endogenous retrovirus (ERV) in the intron 1 of TLR6 was shown to act as an enhancer increasing the activities of TLR6 putative promoter and two mini-promoters. Furthermore, real-time quantitative polymerase chain reaction (qPCR) analysis revealed significant association (p < 0.05) of the ERV insertion with increased mRNA expression of TLR6, the neighboring gene TLR1, and genes downstream in the TLR signaling pathway such as MyD88 (Myeloid differentiation factor 88), Rac1 (Rac family small GTPase 1), TIRAP (TIR domain containing adaptor protein), Tollip (Toll interacting protein) as well as the inflammatory factors IL6 (Interleukin 6), IL8 (Interleukin 8), and TNFα (Tumor necrosis factor alpha) in tissues of 30 day-old piglet. In addition, serum IL6 and TNFα concentrations were also significantly upregulated by the ERV insertion (p < 0.05). Conclusions A total of five RIPs were identified in five different TLR loci. The 192 bp ERV insertion in the first intron of TLR6 was associated with higher expression of TLR6, TLR1, and several genes downstream in the signaling cascade. Thus, the ERV insertion may act as an enhancer affecting regulation of the TLR signaling pathways, and can be potentially applied in breeding of disease resistant animals. Supplementary Information The online version contains supplementary material available at 10.1186/s13100-021-00248-w.
Collapse
Affiliation(s)
- XiaoYan Wang
- College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Zixuan Chen
- College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Eduard Murani
- Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Enrico D'Alessandro
- Department of Veterinary Science, Unit of Animal Production, University of Messina, 98168, Messina, Italy
| | - Yalong An
- College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Cai Chen
- College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Kui Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, 100081, Beijing, China
| | - Grazia Galeano
- Department of Veterinary Science, Unit of Animal Production, University of Messina, 98168, Messina, Italy
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Chengyi Song
- College of Animal Science & Technology, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
17
|
Pancreatic Cancer and Immunotherapy: A Clinical Overview. Cancers (Basel) 2021; 13:cancers13164138. [PMID: 34439292 PMCID: PMC8393975 DOI: 10.3390/cancers13164138] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with high mortality. The vast majority of patients present with unresectable, advanced stage disease, for whom standard of care chemo(radio)therapy may improve survival by several months. Immunotherapy has led to a fundamental shift in the treatment of several advanced cancers. However, its efficacy in PDAC in terms of clinical benefit is limited, possibly owing to the immunosuppressive, inaccessible tumor microenvironment. Still, various immunotherapies have demonstrated the capacity to initiate local and systemic immune responses, suggesting an immune potentiating effect. In this review, we address PDAC's immunosuppressive tumor microenvironment and immune evasion methods and discuss a wide range of immunotherapies, including immunomodulators (i.e., immune checkpoint inhibitors, immune stimulatory agonists, cytokines and adjuvants), oncolytic viruses, adoptive cell therapies (i.e., T cells and natural killer cells) and cancer vaccines. We provide a general introduction to their working mechanism as well as evidence of their clinical efficacy and immune potentiating abilities in PDAC. The key to successful implementation of immunotherapy in this disease may rely on exploitation of synergistic effects between treatment combinations. Accordingly, future treatment approaches should aim to incorporate diverse and novel immunotherapeutic strategies coupled with cytotoxic drugs and/or local ablative treatment, targeting a wide array of tumor-induced immune escape mechanisms.
Collapse
|
18
|
Kaur D, Arora C, Raghava GPS. Prognostic Biomarker-Based Identification of Drugs for Managing the Treatment of Endometrial Cancer. Mol Diagn Ther 2021; 25:629-646. [PMID: 34155607 DOI: 10.1007/s40291-021-00539-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Uterine corpus endometrial carcinoma (UCEC) causes thousands of deaths per year. To improve the overall survival of patients with UCEC, there is a need to identify prognostic biomarkers and potential drugs. OBJECTIVES The aim of this study was twofold: the identification of prognostic gene signatures from expression profiles of pattern recognition receptor (PRR) genes and identification of the most effective existing drugs using the prognostic gene signature. METHODS This study was based on the expression profile of PRR genes of 541 patients with UCEC obtained from The Cancer Genome Atlas. Key prognostic signatures were identified using various approaches, including survival analysis, network, and clustering. Hub genes were identified by constructing a co-expression network. Representative genes were identified using k-means and k-medoids-based clustering. Univariate Cox proportional hazard (PH) analysis was used to identify survival-associated genes. 'cmap2' was used to identify potential drugs that can suppress/enhance the expression of prognostic genes. RESULTS Models were developed using hub genes and achieved a maximum hazard ratio (HR) of 1.37 (p = 0.294). Then, a clustering-based model was developed using seven genes (HR 9.14; p = 1.49 × 10-12). Finally, a nine gene-based risk stratification model was developed (CLEC1B, CLEC3A, IRF7, CTSB, FCN1, RIPK2, NLRP10, NLRP9, and SARM1) and achieved HR 10.70; p = 1.1 × 10-12. The performance of this model improved significantly in combination with the clinical stage and achieved HR 15.23; p = 2.21 × 10-7. We also developed a model for predicting high-risk patients (survival ≤ 4.3 years) and achieved an area under the receiver operating characteristic curve (AUROC) of 0.86. CONCLUSION We identified potential immunotherapeutic agents based on prognostic gene signature: hexamethonium bromide and isoflupredone. Several novel candidate drugs were suggested, including human interferon-α-2b, paclitaxel, imiquimod, MESO-DAP1, and mifamurtide. These biomolecules and repurposed drugs may be utilised for prognosis and treatment for better survival.
Collapse
Affiliation(s)
- Dilraj Kaur
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi, Okhla Industrial Estate, New Delhi, 110020, India
| | - Chakit Arora
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi, Okhla Industrial Estate, New Delhi, 110020, India
| | - Gajendra Pal Singh Raghava
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi, Okhla Industrial Estate, New Delhi, 110020, India.
| |
Collapse
|
19
|
Liu Y, Jin ZR, Huang X, Che YC, Liu Q. Identification of Spindle and Kinetochore-Associated Family Genes as Therapeutic Targets and Prognostic Biomarkers in Pancreas Ductal Adenocarcinoma Microenvironment. Front Oncol 2020; 10:553536. [PMID: 33224872 PMCID: PMC7667267 DOI: 10.3389/fonc.2020.553536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 09/08/2020] [Indexed: 12/16/2022] Open
Abstract
Aim The role of spindle and kinetochore-associated (SKA) genes in tumorigenesis and cancer progression has been widely studied. However, so far, the oncogenic involvement of SKA family genes in pancreatic cancer and their prognostic potential remain unknown. Methods Here, we carried out a meta-analysis of the differential expression of SKA genes in normal and tumor tissue. Univariate and multivariate survival analyses were done to evaluate the correlation between SKA family gene expression and pancreas ductal adenocarcinoma (PDAC) prognosis. Joint-effect and stratified survival analysis as well as nomogram analysis were used to estimate the prognostic value of genes. The underlying regulatory and biological mechanisms were identified by Gene set enrichment analysis. Interaction between SKA prognosis-related genes and immune cell infiltration was assessed using the Tumor Immune Estimation Resource tool. Results We find that SKA1-3 are highly expressed in PDAC tissues relative to non-cancer tissues. Survival analysis revealed that high expression of SKA1 and SKA3 independently indicate poor prognosis but they are not associated with relapse-free survival. The prognostic value of SKA1 and SKA3 was further confirmed by the nomogram, joint-effect, and stratified survival analysis. Analysis of underlying mechanisms reveals that these genes influence cancer-related signaling pathways, kinases, miRNA, and E2F family genes. Notably, prognosis-related genes are inversely correlated with several immune cells infiltrating levels. Conclusion We find that SKA1 and SKA3 expression correlates with prognosis and immune cell infiltration in PDAC, highlighting their potential as pancreatic cancer prognostic biomarkers.
Collapse
Affiliation(s)
- Yi Liu
- Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Guangxi Clinical Research Center for Colorectal Cancer, Nanning, China
| | - Zong-Rui Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xing Huang
- Department of Radiotherapy, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ye-Cheng Che
- Department of Emergency Medicine, First People's Hospital of Fuzhou, Fuzhou, China
| | - Qin Liu
- Department of Medical Ultrasonics, Second People's Hospital of Guilin, Guilin, China
| |
Collapse
|
20
|
Fan L, Zhou P, Chen AX, Liu GY, Yu KD, Shao ZM. Toll-like receptor 3 -926T>A increased the risk of breast cancer through decreased transcriptional activity. Oncoimmunology 2019; 8:e1673126. [PMID: 31741776 PMCID: PMC6844318 DOI: 10.1080/2162402x.2019.1673126] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/22/2019] [Accepted: 09/24/2019] [Indexed: 12/23/2022] Open
Abstract
Toll-like receptor 3 (TLR3) is a viral sensor that induces apoptosis in response to double-stranded RNA (dsRNA). Common genetic changes in the TLR3 gene may influence breast cancer susceptibility and development. However, all of the polymorphisms in the previous study were only markers of the TLR3 gene, not causative polymorphisms. In this study, we performed a case-control study focusing on the relationship between rs5743305 (-926T>A), a single nucleotide polymorphism (SNP) in the promoter region of TLR3, and breast cancer. We found that the genetic variant rs5743305 increased the risk of breast cancer under the dominant and codominant models (dominant model: AT+AA vs TT.: OR = 1.3023, 95%CI: 1.0778-1.5736, P = .0062; codominant model: AA vs. TT: OR = 1.3919, 95%CI: 1.0177-1.9036, P = .0384; AT vs. TT: OR = 1.2799, 95%CI: 1.0475-1.5639, P = .0158) but not under the recessive model (TT vs. AT+AA, OR = 1.2387, 95%CI: 0.9197-1.6682, P = .1588). The same trends were found in the age-adjusted logistic regression study and stage 2 study. Furthermore, the electrophoretic mobility shift assay (EMSA) and luciferase reporter assay showed that rs5743305 decreased the transcriptional activity of TLR3. There was consistently reduced TLR3 mRNA and protein expression in human breast cancer samples from patients with TLR3 - 926A. Therefore, TLR3 rs5743305 increases the risk of breast cancer by decreasing the transcriptional activity of TLR3. This study may provide a better understanding of the genetic architecture underlying disease susceptibility and may advance the potential for preclinical prediction in future genetic testing.
Collapse
Affiliation(s)
- Lei Fan
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Peng Zhou
- Parkway Health, Shanghai, P.R. China
| | - Ao-Xiang Chen
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Guang-Yu Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| | - Zhi-Ming Shao
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, P.R. China
| |
Collapse
|