1
|
Wadhonkar K, Das S, Subramanian R, Sk MH, Singh Y, Baig MS. The effect of cancer cell-derived exosomal proteins on macrophage polarization: An in-depth review. Exp Cell Res 2025; 444:114393. [PMID: 39710293 DOI: 10.1016/j.yexcr.2024.114393] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 12/24/2024]
Abstract
Cancer is characterized by unregulated cell proliferation, enabling it to invade and spread to different organs and tissues in the body. Cancer progression is intricately influenced by the complex dynamics within the tumor microenvironment (TME). The TME is a composite and dynamic network comprising cancer cells and various immune cells, including tumor-associated macrophages. Exosomes facilitate the communication between different cancer cells as well as other types of cells. This review particularly focuses on exosomal proteins derived from different cancer cells in mounting the complex crosstalk between cells of cancer and macrophages within the TME. Most cancer-derived exosomal proteins polarize macrophages towards M2 phenotype, promoting cancer aggressiveness, while a few have role switching towards the M1 phenotype, inhibiting cancer proliferation, respectively. In this review, we summarize, for the first time, the dual impact of cancer cell-derived exosomal proteins on macrophage polarization and the associated signaling pathways, offering valuable insights for developing innovative therapeutic strategies against diverse cancer types.
Collapse
Affiliation(s)
- Khandu Wadhonkar
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Soumalya Das
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | | | - Mobbassar Hassan Sk
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK; Institute for Energy and Environmental Flows, University of Cambridge, Cambridge, UK
| | - Yashi Singh
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India.
| |
Collapse
|
2
|
Chen W, Zhang L, Gao M, Zhang N, Wang R, Liu Y, Niu Y, Jia L. Role of tertiary lymphoid structures and B cells in clinical immunotherapy of gastric cancer. Front Immunol 2025; 15:1519034. [PMID: 39840050 PMCID: PMC11747648 DOI: 10.3389/fimmu.2024.1519034] [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: 10/29/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Gastric cancer is a common malignant tumor of the digestive tract, and its treatment remains a significant challenge. In recent years, the role of various immune cells in the tumor microenvironment in cancer progression and treatment has gained increasing attention. Immunotherapy, primarily based on immune checkpoint inhibitors, has notably improved the prognosis of patients with gastric cancer; however, challenges regarding therapeutic efficacy persist. Histological features within the tumor microenvironment, such as tertiary lymphoid structures (TLSs), tumor-infiltrating lymphocytes, and the proportion of intratumoral stroma, are emerging as potentially effective prognostic factors. In gastric cancer, TLSs may serve as local immune hubs, enhancing the ability of immune cells to interact with and recognize tumor antigens, which is closely linked to the effectiveness of immunotherapy and improved survival rates in patients. However, the specific cell type driving TLS formation in tumors has not yet been elucidated. Mature TLSs are B-cell regions containing germinal centers. During germinal center formation, B cells undergo transformations to become mature cells with immune function, exerting anti-tumor effects. Therefore, targeting B cells within TLSs could provide new avenues for gastric cancer immunotherapy. This review, combined with current research on TLSs and B cells in gastric cancer, elaborates on the relationship between TLSs and B cells in the prognosis and immunotherapy of patients with gastric cancer, aiming to provide effective guidance for precise immunotherapy.
Collapse
Affiliation(s)
- Weiyi Chen
- Basic Medical Sciences, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lingli Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Man Gao
- Bayannur Clinical Medical College, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Ning Zhang
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Rumeng Wang
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Yang Liu
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
| | - Yan Niu
- Medical Experiment Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Lizhou Jia
- Central Laboratory, Bayannur Hospital, Bayannur, Inner Mongolia, China
- Medical Experiment Center, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| |
Collapse
|
3
|
Zaroon, Aslam S, Hafsa, Mustafa U, Fatima S, Bashir H. Interleukin in Immune-Mediated Diseases: An Updated Review. Mol Biotechnol 2024:10.1007/s12033-024-01347-8. [PMID: 39715931 DOI: 10.1007/s12033-024-01347-8] [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: 08/20/2024] [Accepted: 11/29/2024] [Indexed: 12/25/2024]
Abstract
The immune system comprises various regulators and effectors that elicit immune responses against various attacks on the body. The pathogenesis of autoimmune diseases is derived from the deregulated expression of cytokines, the major regulators of the immune system. Among cytokines, interleukins have a major influence on immune-mediated diseases. These interleukins initiate the immune response against healthy and normal cells of the body, resulting in immune-mediated disease. The major interleukins in this respect are IL-1, IL-3, IL-4, IL-6, IL-10 and IL-12 which cause immune responses such as excessive inflammation, loss of immune tolerance, altered T-cell differentiation, immune suppression dysfunction, and inflammatory cell recruitment. Systemic Lupus Erythematosus (SLE) is an autoimmune illness characterized by dysregulation of interleukins. These immune responses are the signs of diseases such as rheumatoid arthritis, inflammatory bowel disease, psoriasis, type I diabetes, and multiple sclerosis.
Collapse
Affiliation(s)
- Zaroon
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Shakira Aslam
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hafsa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Usama Mustafa
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Sana Fatima
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Hamid Bashir
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan.
| |
Collapse
|
4
|
Jabbarzadeh Kaboli P, Roozitalab G, Farghadani R, Eskandarian Z, Zerrouqi A. c-MET and the immunological landscape of cancer: novel therapeutic strategies for enhanced anti-tumor immunity. Front Immunol 2024; 15:1498391. [PMID: 39664377 PMCID: PMC11632105 DOI: 10.3389/fimmu.2024.1498391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024] Open
Abstract
Cellular mesenchymal-epithelial transition factor (c-MET), also known as hepatocyte growth factor receptor (HGFR), is a crucial receptor tyrosine kinase implicated in various solid tumors, including lung, breast, and liver cancers. The concomitant expression of c-MET and PD-L1 in tumors, such as hepatocellular carcinoma, highlights their prognostic significance and connection to therapeutic resistance. Cancer-associated fibroblasts and mesenchymal stromal cells produce hepatocyte growth factor (HGF), activating c-MET signaling in tumor cells and myeloid-derived suppressor cells (MDSC). This activation leads to metabolic reprogramming and increased activity of enzymes like glutaminase (GLS), indoleamine 2,3-dioxygenase (IDO), and arginase 1 (ARG1), depleting essential amino acids in the tumor microenvironment that are vital for effector immune cell function. This review highlights the interplay between tumor cells and myeloid-derived suppressor cells (MDSCs) that create an immunosuppressive environment while providing targets for c-MET-focused immunotherapy. It emphasizes the clinical implications of c-MET inhibition on the behavior of immune cells such as neutrophils, macrophages, T cells, and NK cells. It explores the potential of c-MET antagonism combined with immunotherapeutic strategies to enhance cancer treatment paradigms. This review also discusses the innovative cancer immunotherapies targeting c-MET, including chimeric antigen receptor (CAR) therapies, monoclonal antibodies, and antibody-drug conjugates, while encouraging the development of a comprehensive strategy that simultaneously tackles immune evasion and enhances anti-tumor efficacy further to improve the clinical prognoses for patients with c-MET-positive malignancies. Despite the challenges and variability in efficacy across different cancer subtypes, continued research into the molecular mechanisms and the development of innovative therapeutic strategies will be crucial.
Collapse
Affiliation(s)
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Zoya Eskandarian
- Research Institute Children’s Cancer Center, and Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Abdessamad Zerrouqi
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
5
|
Hu L, Wang X, Song Z, Chen F, Wu B. Leveraging CAR macrophages targeting c-Met for precision immunotherapy in pancreatic cancer: insights from single-cell multi-omics. Mol Med 2024; 30:231. [PMID: 39592929 PMCID: PMC11590533 DOI: 10.1186/s10020-024-00996-4] [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: 08/27/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Pancreatic cancer is known for its poor prognosis and resistance to conventional therapies, largely due to the presence of cancer stem cells (CSCs) and aggressive angiogenesis. Effectively targeting these CSCs and associated angiogenic pathways is crucial for effective treatment. This study leverages single-cell multi-omics to explore a novel therapeutic approach involving Chimeric Antigen Receptor (CAR) macrophages engineered to target the c-Met protein on pancreatic CSCs. METHODS We employed single-cell RNA sequencing to analyze pancreatic cancer tissue, identifying c-Met as a key marker of CSCs. CAR macrophages were engineered using a lentiviral system to express a c-Met-specific receptor. The phagocytic efficiency of these CAR macrophages against pancreatic CSCs was assessed in vitro, along with their ability to inhibit angiogenesis. The in vivo efficacy of CAR macrophages was evaluated in a mouse model of pancreatic cancer. RESULTS CAR macrophages demonstrated high specificity for c-Met + CSCs, significantly enhancing phagocytosis and reducing the secretion of angiogenic factors such as VEGFA, FGF2, and ANGPT. In vivo, these macrophages significantly suppressed tumor growth and angiogenesis, prolonging survival in pancreatic cancer-bearing mice. CONCLUSION CAR macrophages targeting c-Met represent a promising therapeutic strategy for pancreatic cancer, offering targeted elimination of CSCs and disruption of tumor angiogenesis. This study highlights the potential of single-cell multi-omics in guiding the development of precision immunotherapies.
Collapse
Affiliation(s)
- Lingyu Hu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Xiaoguang Wang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China
| | - Bin Wu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, No. 1518 North Huancheng Road, Jiaxing, Zhejiang, 314000, People's Republic of China.
| |
Collapse
|
6
|
Sun J, Corradini S, Azab F, Shokeen M, Muz B, Miari KE, Maksimos M, Diedrich C, Asare O, Alhallak K, Park C, Lubben B, Chen Y, Adebayo O, Bash H, Kelley S, Fiala M, Bender DE, Zhou H, Wang S, Vij R, Williams MTS, Azab AK. IL-10R inhibition reprograms tumor-associated macrophages and reverses drug resistance in multiple myeloma. Leukemia 2024; 38:2355-2365. [PMID: 39215060 PMCID: PMC11518999 DOI: 10.1038/s41375-024-02391-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 07/19/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Multiple myeloma (MM) is the cancer of plasma cells within the bone marrow and remains incurable. Tumor-associated macrophages (TAMs) within the tumor microenvironment often display a pro-tumor phenotype and correlate with tumor proliferation, survival, and therapy resistance. IL-10 is a key immunosuppressive cytokine that leads to recruitment and development of TAMs. In this study, we investigated the role of IL-10 in MM TAM development as well as the therapeutic application of IL-10/IL-10R/STAT3 signaling inhibition. We demonstrated that IL-10 is overexpressed in MM BM and mediates M2-like polarization of TAMs in patient BM, 3D co-cultures in vitro, and mouse models. In turn, TAMs promote MM proliferation and drug resistance, both in vitro and in vivo. Moreover, inhibition of IL-10/IL-10R/STAT3 axis using a blocking IL-10R monoclonal antibody and STAT3 protein degrader/PROTAC prevented M2 polarization of TAMs and the consequent TAM-induced proliferation of MM, and re-sensitized MM to therapy, in vitro and in vivo. Therefore, our findings suggest that inhibition of IL-10/IL-10R/STAT3 axis is a novel therapeutic strategy with monotherapy efficacy and can be further combined with current anti-MM therapy, such as immunomodulatory drugs, to overcome drug resistance. Future investigation is warranted to evaluate the potential of such therapy in MM patients.
Collapse
Affiliation(s)
- Jennifer Sun
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO, USA
| | - Stefan Corradini
- Charles Oakley Laboratories, Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Feda Azab
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Monica Shokeen
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO, USA
- Department of Radiology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Katerina E Miari
- Charles Oakley Laboratories, Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Mina Maksimos
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Camila Diedrich
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Obed Asare
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO, USA
| | - Chaelee Park
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Berit Lubben
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Yixuan Chen
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Ola Adebayo
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Hannah Bash
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sarah Kelley
- Department of Medicine, Oncology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Mark Fiala
- Department of Medicine, Oncology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Diane E Bender
- Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO, USA
| | - Haibin Zhou
- Department of Internal Medicine University of Michigan, Ann Arbor, Michigan, USA
| | - Shaomeng Wang
- Department of Internal Medicine University of Michigan, Ann Arbor, Michigan, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, USA
| | - Ravi Vij
- Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO, USA
- Department of Medicine, Oncology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Mark T S Williams
- Charles Oakley Laboratories, Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow, UK
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Cancer Biology Division, Washington University in St. Louis School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis McKelvey School of Engineering, St. Louis, MO, USA.
- Department of Biomedical Engineering, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO, USA.
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
7
|
Cheng YC, Acedera JD, Li YJ, Shieh SY. A keratinocyte-adipocyte signaling loop is reprogrammed by loss of BTG3 to augment skin carcinogenesis. Cell Death Differ 2024; 31:970-982. [PMID: 38714880 PMCID: PMC11303697 DOI: 10.1038/s41418-024-01304-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 08/09/2024] Open
Abstract
Obesity is endemic to many developed countries. Overweight or obesity is associated with an increased risk of developing cancer. Dysfunctional adipose tissue alters cancer cell proliferation and migration; however, whether and how neoplastic epithelial cells communicate with adipose tissue and the underlying mechanism are less clear. BTG3 is a member of the anti-proliferative BTG/Tob family and functions as a tumor suppressor. Here, we demonstrated that BTG3 levels are downregulated in basal cell carcinoma and squamous cell carcinoma compared to normal skin tissue, and Btg3 knockout in mice augmented the development of papilloma in a mouse model of DMBA/TPA-induced skin carcinogenesis. Mechanistically, BTG3-knockout keratinocytes promoted adipocyte differentiation mainly through the release of IL1α, IL10, and CCL4, as a result of elevated NF-κB activity. These adipocytes produced CCL20 and FGF7 in a feedback loop to promote keratinocyte migration. Thus, our findings showcased the role of BTG3 in guarding the interplay between keratinocytes and adjacent adipocytes, and identified the underlying neoplastic molecular mediators that may serve as possible targets in the treatment of skin cancer.
Collapse
Affiliation(s)
- Yu-Che Cheng
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jack Dalit Acedera
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Yi-Ju Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Sheau-Yann Shieh
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan.
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
8
|
Hu X, Xie S, Yi X, Ouyang Y, Zhao W, Yang Z, Zhang Z, Wang L, Huang X, Peng M, Yu F. Bidirectional Mendelian Randomization of Causal Relationship between Inflammatory Cytokines and Different Pathological Types of Lung Cancer. J Cancer 2024; 15:4969-4984. [PMID: 39132165 PMCID: PMC11310887 DOI: 10.7150/jca.98301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/07/2024] [Indexed: 08/13/2024] Open
Abstract
Prior research has proposed a potential association between lung cancer and inflammatory cytokines, yet the specific causal relationship remains unclear, especially across various lung cancer pathologies. This study utilized bidirectional Mendelian randomization (MR) to explore these causal connections, unveiling novel insights. Our research revealed distinctive inflammatory cytokine profiles for each subtype of lung cancer and identified potential biomarkers that could refine diagnostic and therapeutic approaches. We applied two-sample Mendelian randomization, leveraging genetic variance data from three extensive genome-wide association studies (GWAS) focusing on different lung cancer types (lung adenocarcinoma: 1590 cases and 314,193 controls of healthy individuals of European descent; lung squamous cell carcinoma: 1510 cases and 314,193 controls of European ancestry; small cell lung cancer: 717 cases and 314,193 controls of European ancestry). A separate GWAS summary on inflammatory cytokines from 8,293 healthy participants was also included. The inverse variance weighting method was utilized to examine causal relationships, with robustness confirmed through multiple sensitivity analyses, including MR-Egger, weighted median, and MR-PRESSO. Our analysis revealed that elevated levels of IL_1RA were associated with an increased risk of lung adenocarcinoma (OR: 1.29, 95% CI: 1.02-1.64, p = 0.031), while higher MCP_1_MCAF levels correlated with a decreased risk of lung squamous cell carcinoma (OR: 0.77, 95% CI: 0.61-0.98, p = 0.031). Furthermore, IL_10, IL_13, and TRAIL levels were positively associated with lung squamous cell carcinoma risk (IL_10: OR: 1.27, 95% CI: 1.06-1.53, p = 0.012; IL_13: OR: 1.15, 95% CI: 1.06-1.53, p = 0.036; TRAIL: OR: 1.15, 95% CI: 1.06-1.53, p = 0.043). No association was found between inflammatory cytokine levels and small cell lung cancer development, whereas SDF_1A and B-NGF were linked to an increased risk of this cancer type (SDF_1A: OR: 1.13, 95% CI: 1.05-1.21, p = 0.001; B-NGF: OR: 1.13, 95% CI: 1.01-1.27, p = 0.029). No significant relationship was observed between the 41 circulating inflammatory cytokines and lung adenocarcinoma or squamous cell carcinoma development. Our findings indicate distinct associations between specific inflammatory cytokines and different types of lung cancer. Elevated IL_1RA levels are a risk marker for lung adenocarcinoma, whereas higher MCP_1_MCAF levels appear protective against lung squamous cell carcinoma. Conversely, elevated levels of IL_10, IL_13, and TRAIL are linked with an increased risk of lung squamous cell carcinoma. The relationships of SDF_1A and B-NGF with small-cell lung cancer highlight the complexity of inflammatory markers in cancer development. This study provides a nuanced understanding of the role of inflammatory cytokines in lung cancer, underscoring their potential in refining diagnosis and treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Muyun Peng
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha 410000, China
| |
Collapse
|
9
|
Feng Z, Gao L, Lu Y, He X, Xie J. The potential contribution of aberrant cathepsin K expression to gastric cancer pathogenesis. Discov Oncol 2024; 15:218. [PMID: 38856944 PMCID: PMC11164852 DOI: 10.1007/s12672-023-00814-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/26/2023] [Indexed: 06/11/2024] Open
Abstract
The role of cathepsin K (CTSK) expression in the pathogenesis and progression of gastric cancer (GC) remains unclear. Hence, the primary objective of this study is to elucidate the precise expression and biological role of CTSK in GC by employing a combination of bioinformatics analysis and in vitro experiments. Our findings indicated a significant upregulation of CTSK in GC. The bioinformatics analysis revealed that GC patients with a high level of CTSK expression exhibited enrichment of hallmark gene sets associated with angiogenesis, epithelial-mesenchymal transition (EMT), inflammatory response, KRAS signaling up, TNFα signaling via KFκB, IL2-STAT5 signaling, and IL6-JAK-STAT3 signaling. Additionally, these patients demonstrated elevated levels of M2-macrophage infiltration, which was also correlated with a poorer prognosis. The results of in vitro experiments provided confirmation that the over-expression of CTSK leads to an increase in the proliferative and invasive abilities of GC cells. However, further evaluation was necessary to determine the impact of CTSK on the migration capability of these cells. Our findings suggested that CTSK has the potential to facilitate the initiation and progression of GC by augmenting the invasive capacity of GC cells, engaging in tumor-associated EMT, and fostering the establishment of an immunosuppressive tumor microenvironment (TME).
Collapse
Affiliation(s)
- Zhijun Feng
- Jiangmen Central Hospital, No. 23, Haibang Street, Pengjiang District, Jiangmen, Guangdong, China
- The Second Clinical Medical College, Lanzhou University, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Lina Gao
- Laboratory Medicine Center, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Yapeng Lu
- Department of Anesthesiology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China
| | - Xiaodong He
- The Second Clinical Medical College, Lanzhou University, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China.
| | - Jianqin Xie
- Department of Anesthesiology, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China.
- The Second Clinical Medical College, Lanzhou University, No. 82, Cuiyingmen, Chengguan District, Lanzhou, Gansu, China.
| |
Collapse
|
10
|
Pradhan R, Kundu A, Kundu CN. The cytokines in tumor microenvironment: from cancer initiation-elongation-progression to metastatic outgrowth. Crit Rev Oncol Hematol 2024; 196:104311. [PMID: 38442808 DOI: 10.1016/j.critrevonc.2024.104311] [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: 09/16/2023] [Revised: 02/07/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
It is a well-known fact that cancer can be augmented by infections and inflammation. In fact, chronic inflammation establishes a tumor-supporting-microenvironment (TME), which contributes to neoplastic progression. Presently, extensive research is going on to establish the interrelationship between infection, inflammation, immune response, and cancer. Cytokines are the most essential components in this linkage, which are secreted by immune cells and stromal cells of TME. Cytokines have potential involvement in tumor initiation, elongation, progression, metastatic outgrowth, angiogenesis, and development of therapeutic resistance. They are also linked with increased cancer symptoms along with reduced quality of life in advanced cancer patients. The cancer patients experience multiple symptoms including pain, asthenia, fatigue, anorexia, cachexia, and neurodegenerative disorders etc. Anti-cancer therapeutics can be developed by targeting cytokines along with TME to reduce the immunocompromised state and also modulate the TME. This review article depicts the composition and function of different inflammatory cells within the TME, more precisely the role of cytokines in cancer initiation, elongation, and progression as well as the clinical effects in advanced cancer patients. It also provides an overview of different natural compounds, nanoparticles, and chemotherapeutic agents that can target cytokines along with TME, which finally pave the way for cytokines-targeted anti-cancer therapeutics.
Collapse
Affiliation(s)
- Rajalaxmi Pradhan
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| | - Anushka Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| | - Chanakya Nath Kundu
- Cancer Biology Division, School of Biotechnology, Kalinga Institute of Industrial Technology, Deemed to be University, Campus-11, Patia, Bhubaneswar, Odisha 751024, India.
| |
Collapse
|
11
|
Chang CC, Yang CH, Chuang CH, Jiang SJ, Hwang YM, Liou JW, Hsu HJ. A peptide derived from interleukin-10 exhibits potential anticancer activity and can facilitate cell targeting of gold nanoparticles loaded with anticancer therapeutics. Commun Chem 2023; 6:278. [PMID: 38102207 PMCID: PMC10724200 DOI: 10.1038/s42004-023-01079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 12/01/2023] [Indexed: 12/17/2023] Open
Abstract
Human interleukin-10 (IL-10) is an immunosuppressive and anti-inflammatory cytokine, and its expression is upregulated in tumor tissues and serum samples of patients with various cancers. Because of its immunosuppressive nature, IL-10 has also been suggested to be a factor leading to tumor cells' evasion of immune surveillance and clearance by the host immune system. In this study, we refined a peptide with 20 amino acids, named NK20a, derived from the binding region of IL-10 on the basis of in silico analysis of the complex structure of IL-10 with IL-10Ra, the ligand binding subunit of the IL-10 receptor. The binding ability of the peptide was confirmed through in vitro biophysical biolayer interferometry and cellular experiments. The IL-10 inhibitory peptide exerted anticancer effects on lymphoma B cells and could abolish the suppression effect of IL-10 on macrophages. NK20a was also conjugated with gold nanoparticles to target the chemotherapeutic 5-fluorouracil (5-FU)-loaded nanoparticles to enhance the anticancer efficacy of 5-FU against the breast cancer cell line BT-474. Our study demonstrated that NK20a designed in silico with improved binding affinity to the IL-10 receptor can be used as a tool in developing anticancer strategies.
Collapse
Affiliation(s)
- Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan, ROC
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Chin-Hao Yang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Chin-Hsien Chuang
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Yin-Min Hwang
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC
| | - Je-Wen Liou
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
| | - Hao-Jen Hsu
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
- Department of Biomedical Sciences and Engineering, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan, ROC.
| |
Collapse
|
12
|
Sivasankar C, Hewawaduge C, Muthuramalingam P, Lee JH. Tumor-targeted delivery of lnc antisense RNA against RCAS1 by live-attenuated tryptophan-auxotrophic Salmonella inhibited 4T1 breast tumors and metastasis in mice. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 34:102053. [PMID: 37941832 PMCID: PMC10628790 DOI: 10.1016/j.omtn.2023.102053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 10/11/2023] [Indexed: 11/10/2023]
Abstract
Emerging chemo- and radiotherapy resistance exacerbated the cancer risk and necessitated novel treatment strategies. Although RNA therapeutics against pro-oncogenic genes are highly effective, tumor-specific delivery remains a barrier to the implementation of this valuable tool. In this study, we report a tryptophan-auxotrophic Salmonella typhimurium strain as an onco-therapeutic delivery system with tumor-targeting ability using 4T1 mice breast-cancer model. The receptor-binding cancer antigen expressed on SiSo cell (RCAS1) is a cancer-specific protein that induces the apoptosis of peripheral lymphocytes and confers tumor immune evasion. We designed a long non-coding antisense-RNA against RCAS1 (asRCAS1) and delivered by Salmonella using a non-antibiotic, auxotrophic-selective, eukaryotic expression plasmid, pJHL204. After in vivo tumor-to-tumor passaging, the JOL2888 (ΔtrpA, ΔtrpE, Δasd + asRCAS1) strain exhibited high sustainability in tumors, but did not last in healthy organs, thereby demonstrating tumor specificity and safety. RCAS1 inhibition in the tumor was confirmed by western blotting and qPCR. In mice, JOL2888 treatment reduced tumor-associated macrophages, improved the T cell population, elicited cell-mediated immunity, and suppressed cancer-promoting genes. Consequently, the JOL2888 treatment significantly decreased the tumor volume by 80%, decreased splenomegaly by 30%, and completely arrested lung metastasis. These findings highlight the intrinsic tumor-targeting ability of tryptophan-auxotrophic Salmonella for delivering onco-therapeutic macromolecules.
Collapse
Affiliation(s)
- Chandran Sivasankar
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | - Chamith Hewawaduge
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| | | | - John Hwa Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan Campus 54596, Republic of Korea
| |
Collapse
|
13
|
Co EL, Hameed M, Sebastian SA, Garg T, Sudan S, Bheemisetty N, Mohan B. Narrative Review of Probiotic Use on the Recovery of Postoperative Patients with Esophageal Cancer. Curr Nutr Rep 2023; 12:635-642. [PMID: 37605086 DOI: 10.1007/s13668-023-00490-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 08/23/2023]
Abstract
PURPOSE OF REVIEW This narrative review discusses the significance of probiotic therapy in the postoperative care of patients with esophageal cancer and its role as an adjunct therapy to other treatment modalities for esophageal cancer. RECENT FINDINGS As such, there is an emerging need to address any malnutrition and gastrointestinal problems occurring in these patients which tend to have a strong negative impact on their prognosis. Probiotic effects on esophageal cancer biomarkers suggest that there is a positive correlation between these two factors. However, the beneficial effects remain controversial and warrant further investigation. Probiotics, now being widely utilized as postoperative therapy in some carcinomas of the gastrointestinal tract such as gastric cancer and colorectal cancer, have been shown in some clinical studies to positively impact the nutritional status of patients with esophageal cancer. Postoperative care among patients suffering from esophageal cancer is a very crucial aspect in the survival of these patients.
Collapse
Affiliation(s)
- Edzel Lorraine Co
- University of Santo Tomas Faculty of Medicine and Surgery, Manila, Philippines
| | - Maha Hameed
- Department of Internal Medicine, Florida State University/Sarasota Memorial Hospital, 1700 S Tamiami Trial, Sarasota, FL, 34239, USA.
| | | | - Tulika Garg
- Government Medical College and Hospital, Chandigarh, India
| | | | | | - Babu Mohan
- Department of Gastroenterology, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
14
|
Jung YJ, Woo JS, Hwang SH, Yang S, Kim SJ, Jhun J, Lee SY, Lee KH, Cho ML, Song KY. Effect of IL-10-producing B cells in peripheral blood and tumor tissue on gastric cancer. Cell Commun Signal 2023; 21:320. [PMID: 37946227 PMCID: PMC10634038 DOI: 10.1186/s12964-023-01174-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/25/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Interleukin (IL)-10-producing B (B10) cells are generated in response to signals from the tumor microenvironment and promote tumor growth by interacting with B10 cells. We investigated the distributions of immune cells in peripheral blood and tumor tissue samples from patients with gastric cancer (GC). METHODS Patients with GC who underwent radical gastrectomy in Seoul St. Mary's Hospital between August 2020 and May 2021 were enrolled in this study. Forty-two samples of peripheral blood were collected, and a pair of gastric mucosal samples (normal and cancerous mucosa; did not influence tumor diagnosis or staging) was collected from each patient after surgery. B10 cells in peripheral blood and cancer mucosa samples were investigated by flow cytometry and immunofluorescence. AGS cells, gastric cancer cell line, were cultured with IL-10 and measured cell death and cytokine secretion. Also, AGS cells were co-cultured with CD19 + B cells and measured cytokine secretion. RESULTS The population of B10 cells was significantly larger in the blood of patients with GC compared with controls. In confocal images of gastric mucosal tissues, cancerous mucosa contained more B10 cells than normal mucosa. The population of B10 cells in cancerous mucosa increased with cancer stage. When AGS cells were cultured under cell-death conditions, cellular necrosis was significantly decreased, and proliferation was increased, for 1 day after IL-10 stimulation. Tumor necrosis factor (TNF)-α, IL-8, IL-1β, and vascular endothelial growth factor secretion by cancer cells was significantly increased by coculture of AGS cells with GC-derived CD19+ B cells. CONCLUSIONS B cells may be one of the populations that promote carcinogenesis by inducing the production of inflammatory mediators, such as IL-10, in GC. Targeting B10 cells activity could improve the outcomes of antitumor immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 07345, Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sun-Hee Hwang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - SeungCheon Yang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - So Jung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
15
|
Carlini V, Noonan DM, Abdalalem E, Goletti D, Sansone C, Calabrone L, Albini A. The multifaceted nature of IL-10: regulation, role in immunological homeostasis and its relevance to cancer, COVID-19 and post-COVID conditions. Front Immunol 2023; 14:1161067. [PMID: 37359549 PMCID: PMC10287165 DOI: 10.3389/fimmu.2023.1161067] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Interleukin-10 (IL-10) is a pleiotropic cytokine that has a fundamental role in modulating inflammation and in maintaining cell homeostasis. It primarily acts as an anti-inflammatory cytokine, protecting the body from an uncontrolled immune response, mostly through the Jak1/Tyk2 and STAT3 signaling pathway. On the other hand, IL-10 can also have immunostimulating functions under certain conditions. Given the pivotal role of IL-10 in immune modulation, this cytokine could have relevant implications in pathologies characterized by hyperinflammatory state, such as cancer, or infectious diseases as in the case of COVID-19 and Post-COVID-19 syndrome. Recent evidence proposed IL-10 as a predictor of severity and mortality for patients with acute or post-acute SARS-CoV-2 infection. In this context, IL-10 can act as an endogenous danger signal, released by tissues undergoing damage in an attempt to protect the organism from harmful hyperinflammation. Pharmacological strategies aimed to potentiate or restore IL-10 immunomodulatory action may represent novel promising avenues to counteract cytokine storm arising from hyperinflammation and effectively mitigate severe complications. Natural bioactive compounds, derived from terrestrial or marine photosynthetic organisms and able to increase IL-10 expression, could represent a useful prevention strategy to curb inflammation through IL-10 elevation and will be discussed here. However, the multifaceted nature of IL-10 has to be taken into account in the attempts to modulate its levels.
Collapse
Affiliation(s)
- Valentina Carlini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Douglas M. Noonan
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Eslam Abdalalem
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Clementina Sansone
- Stazione Zoologica Anton Dohrn, Istituto Nazionale di Biologia, Ecologia e Biotecnologie Marine, Napoli, Italy
| | - Luana Calabrone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), MultiMedica, Milan, Italy
| | - Adriana Albini
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) European Institute of Oncology IEO-, Milan, Italy
| |
Collapse
|
16
|
Zhao Z, Hao Z, Zhang Z, Zhan X. Bioinformatics Analysis Reveals the Vital Role of AKR1B1 in Immune Infiltration and Clinical Outcomes of Gastric Cancer. DNA Cell Biol 2023. [PMID: 37285280 DOI: 10.1089/dna.2022.0644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Infiltrated immune cells are an important constitute of tumor microenvironment, which exert complex effects on gastric cancer (GC) pathogenesis and progression. By using weighted gene co-expression network analysis, integrating the data from The Cancer Genome Atlas-stomach adenocarcinoma and GSE62254, we identify Aldo-Keto Reductase Family 1 Member B (AKR1B1) as a hub gene for immune regulation in GC. Notably, AKR1B1 is associated with higher immune infiltration and worse histologic grade of GC. In addition, AKR1B1 is an independent factor for predicting the survival rate of GC patients. In vitro experiments further demonstrated that AKR1B1-overexpressed THP-1-derived macrophages promoted the proliferation and migration of GC cells. Taken together, AKR1B1 plays an important role in GC progression by regulating immune microenvironment, which could be a biomarker for predicting GC prognosis as well as a potential therapeutic target for GC treatment.
Collapse
Affiliation(s)
- Zhiyue Zhao
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Oncology, Jinling Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Zheng Zhang
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- Department of Orthopedic Rehabilitation, Qingdao Special Servicemen Recuperation Center of PLA Navy, Qingdao, China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| |
Collapse
|
17
|
Lee KH, Kim SJ, Woo JS, Lee SY, Jhun J, Moon J, Jung YJ, Cho ML, Song KY. Prognostic significances of PD-L1- and CTLA-4-positive T cells and positive correlations of immunosuppressive marker expression between cancer tissue and peripheral blood in patients with gastric cancer. Front Immunol 2023; 14:1138743. [PMID: 37153541 PMCID: PMC10160473 DOI: 10.3389/fimmu.2023.1138743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Although tumor, node, metastasis (TNM) staging has been used for prognostic assessment of gastric cancer (GC), the prognosis may vary among patients with the same TNM stage. Recently, the TNM-Immune (TNM-I) classification staging system has been used for prognostic assessment of colorectal cancer based on intra-tumor T-cell status, which is a superior prognostic factor compared with the American Joint Committee on Cancer staging manual. However, an immunoscoring system with prognostic significance for GC has not been established. Method Here, we evaluated immune phenotypes in cancer and normal tissues, then examined correlations between tissues and peripheral blood. GC patients who underwent gastrectomy at Seoul St. Mary's Hospital between February 2000 and May 2021 were included. We collected 43 peripheral blood samples preoperatively and a pair of gastric mucosal samples postoperatively, including normal and cancer mucosa, which did not influence tumor diagnosis and staging. Tissue microarray samples of GC were collected from 136 patients during surgery. We investigated correlations of immune phenotypes between tissues and peripheral blood using immunofluorescence imaging and flow cytometry, respectively. GC mucosa exhibited an increased number of CD4+ T cells, as well as increased expression levels of immunosuppressive markers (e.g., programmed death-ligand-1 [PD-L1], cytotoxic T lymphocyte antigen-4 [CTLA-4], and interleukin-10), in CD4+ T cells and non-T cells. Result The expression levels of immunosuppressive markers were significantly increased in cancer tissues and peripheral blood mononuclear cells. In gastric mucosal tissues and peripheral blood of GC patients, similar immunosuppression phenotypes were observed, including increased numbers of PD-L1- and CTLA-4-positive T cells. Discussion Therefore, peripheral blood analysis may be an important tool for prognostic assessment of GC patients.
Collapse
Affiliation(s)
- Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - So Jung Kim
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung Yoon Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jooyeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeonghyeon Moon
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Yoon Ju Jung
- Division of Gastrointestinal Surgery, Department of Surgery, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Biomedicine and Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- *Correspondence: Mi-La Cho, ; Kyo Young Song,
| | - Kyo Young Song
- Division of Gastrointestinal Surgery, Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- *Correspondence: Mi-La Cho, ; Kyo Young Song,
| |
Collapse
|
18
|
Jacenik D, Lebish EJ, Beswick EJ. MK2 Promotes the Development and Progression of Pancreatic Neuroendocrine Tumors Mediated by Macrophages and Metabolomic Factors. Int J Mol Sci 2022; 23:13561. [PMID: 36362348 PMCID: PMC9658113 DOI: 10.3390/ijms232113561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 09/24/2023] Open
Abstract
Cases of pancreatic neuroendocrine tumors (PNETs) are growing in number, and new treatment options are needed in order to improve patient outcomes. The mitogen-activated protein kinase-activated protein kinase 2 (MK2) is a crucial regulator of cytokine/chemokine production. The significance of MK2 expression and signaling pathway mediated by MK2 in PNETs has not been investigated. To characterize the impact of MK2 on PNET growth, we used the RipTag2 transgenic murine model of PNETs, and we developed a primary PNET cell line for both in vitro and in vivo studies. In the transgenic murine model of PNETs, we found that MK2 inhibition improves survival of mice and prevents PNET progression. MK2 blockade abolished cytokine/chemokine production, which was related to macrophage function. A role for MK2 in the regulation of metabolic factor secretion in PNETs was identified, making this the first study to identify a potential role for the MK2 pathway in regulation of tumor metabolism. Moreover, using an in vitro approach and allograft model of PNETs, we were able to show that macrophages with MK2 depletion exhibit increased cytotoxicity against PNET cells and substantially decreased production of pro-inflammatory cytokines and chemokines, as well as metabolic factors. Taken together, our work identifies MK2 as a potent driver of immune response and metabolic effectors in PNETs, suggesting it is a potential therapeutic target for patients with PNETs.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Eric J. Lebish
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Ellen J. Beswick
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
19
|
McRitchie BR, Akkaya B. Exhaust the exhausters: Targeting regulatory T cells in the tumor microenvironment. Front Immunol 2022; 13:940052. [PMID: 36248808 PMCID: PMC9562032 DOI: 10.3389/fimmu.2022.940052] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022] Open
Abstract
The concept of cancer immunotherapy has gained immense momentum over the recent years. The advancements in checkpoint blockade have led to a notable progress in treating a plethora of cancer types. However, these approaches also appear to have stalled due to factors such as individuals' genetic make-up, resistant tumor sub-types and immune related adverse events (irAE). While the major focus of immunotherapies has largely been alleviating the cell-intrinsic defects of CD8+ T cells in the tumor microenvironment (TME), amending the relationship between tumor specific CD4+ T cells and CD8+ T cells has started driving attention as well. A major roadblock to improve the cross-talk between CD4+ T cells and CD8+ T cells is the immune suppressive action of tumor infiltrating T regulatory (Treg) cells. Despite their indispensable in protecting tissues against autoimmune threats, Tregs have also been under scrutiny for helping tumors thrive. This review addresses how Tregs establish themselves at the TME and suppress anti-tumor immunity. Particularly, we delve into factors that promote Treg migration into tumor tissue and discuss the unique cellular and humoral composition of TME that aids survival, differentiation and function of intratumoral Tregs. Furthermore, we summarize the potential suppression mechanisms used by intratumoral Tregs and discuss ways to target those to ultimately guide new immunotherapies.
Collapse
Affiliation(s)
- Bayley R. McRitchie
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Billur Akkaya
- Department of Neurology, The College of Medicine, The Ohio State University, Columbus, OH, United States
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- Department of Microbial Infection and Immunity, The College of Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
20
|
Ma X, Ou K, Liu X, Yang L. Application progress of liquid biopsy in gastric cancer. Front Oncol 2022; 12:969866. [PMID: 36185234 PMCID: PMC9521037 DOI: 10.3389/fonc.2022.969866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors globally. Guiding the individualized treatment of GC is the focus of research. Obtaining representative biological samples to study the biological characteristics of GC is the focus of diagnosis and treatment of GC. Liquid biopsy technology can use high-throughput sequencing technology to detect biological genetic information in blood. Compared with traditional tissue biopsy, liquid biopsy can determine the dynamic changes of tumor. As a noninvasive auxiliary diagnostic method, liquid biopsy can provide diagnostic and prognostic information concerning the progression of the disease. Liquid biopsy includes circulating tumor cells, circulating tumor DNA, circulating tumor RNA, tumor educated platelets, exosomes, and cytokines. This article describes the classification of liquid biopsy and its application value in the occurrence, development, and therapeutic efficacy of GC.
Collapse
|
21
|
Majewski M, Mertowska P, Mertowski S, Smolak K, Grywalska E, Torres K. Microbiota and the Immune System-Actors in the Gastric Cancer Story. Cancers (Basel) 2022; 14:cancers14153832. [PMID: 35954495 PMCID: PMC9367521 DOI: 10.3390/cancers14153832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Stomach cancer is one of the most commonly diagnosed cancers in the world. Although the number of new cases is decreasing year by year, the death rate for this type of cancer is still high. The heterogeneous course and the lack of symptoms in the early stages of the disease mean that the diagnosis is made late, which translates into a worse prognosis for such patients. That is why it is so important to analyze potential risk factors that may increase the risk of developing gastric cancer and to search for new effective methods of treatment. These requirements are met by the analysis of the composition of the gastric microbiota and its relationship with the immune system, which is a key element in the human anti-cancer fight. This publication was created to systematize the current knowledge on the impact of dysbiosis of human microbiota on the development and progression of gastric cancer. Particular emphasis was placed on taking into account the role of the immune system in this process. Abstract Gastric cancer remains one of the most commonly diagnosed cancers in the world, with a relatively high mortality rate. Due to the heterogeneous course of the disease, its diagnosis and treatment are limited and difficult, and it is associated with a reduced prognosis for patients. That is why it is so important to understand the mechanisms underlying the development and progression of this cancer, with particular emphasis on the role of risk factors. According to the literature data, risk factors include: changes in the composition of the stomach and intestinal microbiota (microbiological dysbiosis and the participation of Helicobacter pylori), improper diet, environmental and genetic factors, and disorders of the body’s immune homeostasis. Therefore, the aim of this review is to systematize the knowledge on the influence of human microbiota dysbiosis on the development and progression of gastric cancer, with particular emphasis on the role of the immune system in this process.
Collapse
Affiliation(s)
- Marek Majewski
- 2nd Department of General, Gastrointestinal Surgery and Surgical Oncology of the Alimentary Tract, Medical University of Lublin, 20-081 Lublin, Poland
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Konrad Smolak
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Kamil Torres
- Chair and Department of Didactics and Medical Simulation, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
22
|
Downregulation of IL-8 and IL-10 by the Activation of Ca2+-Activated K+ Channel KCa3.1 in THP-1-Derived M2 Macrophages. Int J Mol Sci 2022; 23:ijms23158603. [PMID: 35955737 PMCID: PMC9368915 DOI: 10.3390/ijms23158603] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/04/2023] Open
Abstract
THP-1-differentiated macrophages are useful for investigating the physiological significance of tumor-associated macrophages (TAMs). In the tumor microenvironment (TME), TAMs with the M2-like phenotype play a critical role in promoting cancer progression and metastasis by inhibiting the immune surveillance system. We examined the involvement of Ca2+-activated K+ channel KCa3.1 in TAMs in expressing pro-tumorigenic cytokines and angiogenic growth factors. In THP-1-derived M2 macrophages, the expression levels of IL-8 and IL-10 were significantly decreased by treatment with the selective KCa3.1 activator, SKA-121, without changes in those of VEGF and TGF-β1. Furthermore, under in vitro experimental conditions that mimic extracellular K+ levels in the TME, IL-8 and IL-10 levels were both significantly elevated, and these increases were reversed by combined treatment with SKA-121. Among several signaling pathways potentially involved in the transcriptional regulation of IL-8 and IL-10, respective treatments with ERK and JNK inhibitors significantly repressed their transcriptions, and treatment with SKA-121 significantly reduced the phosphorylated ERK, JNK, c-Jun, and CREB levels. These results strongly suggest that the KCa3.1 activator may suppress IL-10-induced tumor immune surveillance escape and IL-8-induced tumorigenicity and metastasis by inhibiting their production from TAMs through ERK-CREB and JNK-c-Jun cascades.
Collapse
|
23
|
Ura B, Capaci V, Aloisio M, Di Lorenzo G, Romano F, Ricci G, Monasta L. A Targeted Proteomics Approach for Screening Serum Biomarkers Observed in the Early Stage of Type I Endometrial Cancer. Biomedicines 2022; 10:1857. [PMID: 36009404 PMCID: PMC9405144 DOI: 10.3390/biomedicines10081857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/22/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy, and it arises in the inner part of the uterus. Identification of serum biomarkers is essential for diagnosing the disease at an early stage. In this study, we selected 44 healthy controls and 44 type I EC at tumor stage 1, and we used the Immuno-oncology panel and the Target 96 Oncology III panel to simultaneously detect the levels of 92 cancer-related proteins in serum, using a proximity extension assay. By applying this methodology, we identified 20 proteins, associated with the outcome at binary logistic regression, with a p-value below 0.01 for the first panel and 24 proteins with a p-value below 0.02 for the second one. The final multivariate logistic regression model, combining proteins from the two panels, generated a model with a sensitivity of 97.67% and a specificity of 83.72%. These results support the use of the proposed algorithm after a validation phase.
Collapse
Affiliation(s)
- Blendi Ura
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Valeria Capaci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Michelangelo Aloisio
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Giovanni Di Lorenzo
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Federico Romano
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| | - Giuseppe Ricci
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34129 Trieste, Italy
| | - Lorenzo Monasta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 34137 Trieste, Italy; (V.C.); (M.A.); (G.D.L.); (F.R.); (G.R.); (L.M.)
| |
Collapse
|
24
|
Janiczek-Polewska M, Szylberg Ł, Malicki J, Marszałek A. Role of Interleukins and New Perspectives in Mechanisms of Resistance to Chemotherapy in Gastric Cancer. Biomedicines 2022; 10:1600. [PMID: 35884907 PMCID: PMC9312950 DOI: 10.3390/biomedicines10071600] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 11/17/2022] Open
Abstract
Gastric cancer (GC) is the fourth most common cancer in the world in terms of incidence and second in terms of mortality. Chemotherapy is the main treatment for GC. The greatest challenge and major cause of GC treatment failure is resistance to chemotherapy. As such, research is ongoing into molecular evaluation, investigating mechanisms, and screening therapeutic targets. Several mechanisms related to both the tumor cells and the tumor microenvironment (TME) are involved in resistance to chemotherapy. TME promotes the secretion of various inflammatory cytokines. Recent studies have revealed that inflammatory cytokines affect not only tumor growth, but also chemoresistance. Cytokines in TME can be detected in blood circulation and TME cells. Inflammatory cytokines could serve as potential biomarkers in the assessment of chemoresistance and influence the management of therapeutics in GC. This review presents recent data concerning research on inflammatory cytokines involved in the mechanisms of chemoresistance and provides new clues in GC treatment.
Collapse
Affiliation(s)
- Marlena Janiczek-Polewska
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
- Department of Clinical Oncology, Greater Poland Cancer Center, 61-866 Poznan, Poland
| | - Łukasz Szylberg
- Department of Perinatology, Gynaecology and Gynaecologic Oncology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland;
- Department of Tumor Pathology and Pathomorphology, Oncology Centrer of Franciszek Łukaszczyk Memorial Hospital, 85-796 Bydgoszcz, Poland
| | - Julian Malicki
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznan, Poland;
| | - Andrzej Marszałek
- Department of Oncologic Pathology, Prophylaxis Poznan University, Medical Sciences and Greater Poland Cancer Center, 61-866 Poznan, Poland;
| |
Collapse
|
25
|
Reactive Oxygen Species Bridge the Gap between Chronic Inflammation and Tumor Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2606928. [PMID: 35799889 PMCID: PMC9256443 DOI: 10.1155/2022/2606928] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
According to numerous animal studies, adverse environmental stimuli, including physical, chemical, and biological factors, can cause low-grade chronic inflammation and subsequent tumor development. Human epidemiological evidence has confirmed the close relationship between chronic inflammation and tumorigenesis. However, the mechanisms driving the development of persistent inflammation toward tumorigenesis remain unclear. In this study, we assess the potential role of reactive oxygen species (ROS) and associated mechanisms in modulating inflammation-induced tumorigenesis. Recent reports have emphasized the cross-talk between oxidative stress and inflammation in many pathological processes. Exposure to carcinogenic environmental hazards may lead to oxidative damage, which further stimulates the infiltration of various types of inflammatory cells. In turn, increased cytokine and chemokine release from inflammatory cells promotes ROS production in chronic lesions, even in the absence of hazardous stimuli. Moreover, ROS not only cause DNA damage but also participate in cell proliferation, differentiation, and apoptosis by modulating several transcription factors and signaling pathways. We summarize how changes in the redox state can trigger the development of chronic inflammatory lesions into tumors. Generally, cancer cells require an appropriate inflammatory microenvironment to support their growth, spread, and metastasis, and ROS may provide the necessary catalyst for inflammation-driven cancer. In conclusion, ROS bridge the gap between chronic inflammation and tumor development; therefore, targeting ROS and inflammation represents a new avenue for the prevention and treatment of cancer.
Collapse
|
26
|
Liu J, Jiao L, Zhong X, Yao W, Du K, Lu S, Wu Y, Ma T, Tong J, Xu M, Jiang W, Wang Y, He M, Xin W, Liu M. Platelet Activating Factor Receptor Exaggerates Microglia-Mediated Microenvironment by IL10-STAT3 Signaling: A Novel Potential Biomarker and Target for Diagnosis and Treatment of Alzheimer's Disease. Front Aging Neurosci 2022; 14:856628. [PMID: 35572136 PMCID: PMC9096237 DOI: 10.3389/fnagi.2022.856628] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/17/2022] Open
Abstract
Background Early diagnosis and effective intervention are the keys to delaying the progression of Alzheimer's Disease (AD). Therefore, we aimed to identify new biomarkers for the early diagnosis of AD through bioinformatic analysis and elucidate the possible underlying mechanisms. Methods and Results GSE1297, GSE63063, and GSE110226 datasets from the GEO database were used to screen the highly differentially expressed genes. We identified a potential biomarker, Platelet activating factor receptor (PTAFR), significantly upregulated in the brain tissue, peripheral blood, and cerebrospinal fluid of AD patients. Furthermore, PTAFR levels in the plasma and brain tissues of APP/PS1 mice were significantly elevated. Simultaneously, PTAFR could mediate the inflammatory responses to exaggerate the microenvironment, particularly mediated by the microglia through the IL10-STAT3 pathway. In addition, PTAFR was a putative target of anti-AD compounds, including EGCG, donepezil, curcumin, memantine, and Huperzine A. Conclusion PTAFR was a potential biomarker for early AD diagnosis and treatment which correlated with the microglia-mediated microenvironment. It is an important putative target for the development of a novel strategy for clinical treatment and drug discovery for AD.
Collapse
Affiliation(s)
- Junxiu Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Linchi Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Xin Zhong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Senxu Lu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yuqiang Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Tianxin Ma
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Junhui Tong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Mingyue Xu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Wenjuan Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China
| | - Yubao Wang
- Liaoning Medical Diagnosis and Treatment Center, Shenyang, China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China,Miao He,
| | - Wei Xin
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,The First Affiliated Hospital of China Medical University, Shenyang, China,Wei Xin,
| | - Mingyan Liu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, China Medical University, Shenyang, China,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Center, China Medical University, Shenyang, China,Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, China Medical University, Shenyang, China,*Correspondence: Mingyan Liu,
| |
Collapse
|
27
|
Wang Y, Fang Y, Zhao F, Gu J, Lv X, Xu R, Zhang B, Fang Z, Li Y. Identification of GGT5 as a Novel Prognostic Biomarker for Gastric Cancer and its Correlation With Immune Cell Infiltration. Front Genet 2022; 13:810292. [PMID: 35368661 PMCID: PMC8971189 DOI: 10.3389/fgene.2022.810292] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer (GC) is a common malignant tumor of the digestive system. Recent studies revealed that high gamma-glutamyl-transferase 5 (GGT5) expression was associated with a poor prognosis of gastric cancer patients. In the present study, we aimed to confirm the expression and prognostic value of GGT5 and its correlation with immune cell infiltration in gastric cancer. First, we compared the differential expression of GGT5 between gastric cancer tissues and normal gastric mucosa in the cancer genome atlas (TCGA) and GEO NCBI databases using the most widely available data. Then, the Kaplan-Meier method, Cox regression, and univariate logistic regression were applied to explore the relationships between GGT5 and clinical characteristics. We also investigated the correlation of GGT5 with immune cell infiltration, immune-related genes, and immune checkpoint genes. Finally, we estimated enrichment of gene ontologies categories and relevant signaling pathways using GO annotations, KEGG, and GSEA pathway data. The results showed that GGT5 was upregulated in gastric cancer tissues compared to normal tissues. High GGT5 expression was significantly associated with T stage, histological type, and histologic grade (p < 0.05). Moreover, gastric cancer patients with high GGT5 expression showed worse 10-years overall survival (p = 0.008) and progression-free intervals (p = 0.006) than those with low GGT5 expression. Multivariate analysis suggested that high expression of GGT5 was an independent risk factor related to the worse overall survival of gastric cancer patients. A nomogram model for predicting the overall survival of GC was constructed and computationally validated. GGT5 expression was positively correlated with the infiltration of natural killer cells, macrophages, and dendritic cells but negatively correlated with Th17 infiltration. Additionally, we found that GGT5 was positively co-expressed with immune-related genes and immune checkpoint genes. Functional analysis revealed that differentially expressed genes relative to GGT5 were mainly involved in the biological processes of immune and inflammatory responses. In conclusion, GGT5 may serve as a promising prognostic biomarker and a potential immunological therapeutic target for GC, since it is associated with immune cell infiltration in the tumor microenvironment.
Collapse
Affiliation(s)
- Yuli Wang
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuan Fang
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Fanchen Zhao
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiefei Gu
- Information Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiang Lv
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rongzhong Xu
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Bo Zhang
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhihong Fang
- Department of Oncology II, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Zhihong Fang, ; Yan Li,
| | - Yan Li
- Department of Oncology I, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Zhihong Fang, ; Yan Li,
| |
Collapse
|
28
|
Li L, Xiong W, Li D, Cao J. Association of Interleukin-10 Polymorphism (rs1800896, rs1800871, and rs1800872) With Breast Cancer Risk: An Updated Meta-Analysis Based on Different Ethnic Groups. Front Genet 2022; 13:829283. [PMID: 35186043 PMCID: PMC8855208 DOI: 10.3389/fgene.2022.829283] [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: 12/05/2021] [Accepted: 01/21/2022] [Indexed: 12/28/2022] Open
Abstract
Background: The interleukin10 (IL-10) gene polymorphisms have been indicated to be associated with breast cancer (BC) risk, but the findings are still controversial. To derive a more precise evaluation, we performed a comprehensive meta-analysis. Methods: A systematic literature search was conducted using PubMed, Embase, CNKI, China biomedical (CBM), and Google Scholar to 29 March 2020. Revman5.3 and Stata 12.0 software analyzed the data, and the strength of the association was identified using the odds ratio (OR) and the corresponding 95% confidence interval (CI). Results: A total of 23 studies (7,250 cancer cases and 7,675 case-free controls) were included in this meta-analysis. The results show that IL-10 gene polymorphisms were significantly correlated with BC risk based on subgroup analysis by ethnicity. The IL-10 rs1800896 polymorphism was significantly associated with the risk of BC in Asians (G vs. A: OR = 0.78, 95% CI = 0.65–0.95, p = 0.01; GG vs. AA: OR = 0.51, 95% CI = 0.31–0.84, p = 0.007; GA vs. AA: OR = 0.6, 95% CI = 0.44–0.81, p = 0.0009; GG + GA vs. AA: OR = 0.6, 95% CI = 0.45–0.81, p = 0.0007); Moreover, an increased BC risk in Asians were also associated with the IL-10 rs1800872 polymorphism (AA vs CC: OR = 0.74, 95% CI = 0.55–0.99, p = 0.04; A vs C: OR = 0.85, 95% CI = 0.74–0.98, p = 0.03). In addition, The IL-10 rs1800871 (CT vs. TT: OR = 1.8, 95% CI = 1.03–3.13, p = 0.04) and rs1800872 polymorphism (A vs C: OR = 0.65, 95% CI 0.43–0.98, p = 0.04) were associated with BC risk in Caucasians. Conclusion: Collectively, this meta-analysis demonstrated that IL-10 rs1800896 and rs1800872 (AA vs. CC; A vs. C) polymorphisms significantly increased the risk of BC in Asians, while the rs1800871 and rs1800872 (A vs. C) were associated with the risk of BC in Caucasians. Therefore, this may provide new ideas for predicting and diagnosing BC susceptibility through the detection of IL-10 gene polymorphism. Systematic Review Registration: [https://www.crd.york.ac.uk/ PROSPERO], identifier [CRD42021266635].
Collapse
Affiliation(s)
- Lijun Li
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Xiong
- The Second Affiliated Hospital, Department of Breast and Thyroid Surgical, Hengyang Medical School, University of South China, Hengyang, China
| | - Donghua Li
- The Second Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, Hengyang, China
| | - Jiangang Cao
- The Affiliated Nanhua Hospital, Clinical Research Institute, Hengyang Medical School, University of South China, Hengyang, China
- *Correspondence: Jiangang Cao,
| |
Collapse
|
29
|
Das SC, Rahman MA, Das Gupta S. In-silico analysis unravels the structural and functional consequences of non-synonymous SNPs in the human IL-10 gene. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00223-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Abstract
Background
Interleukin-10 (IL-10) is an anti-inflammatory cytokine that affects different immune cells. It is also associated with the stimulation of the T and B cells for the production of antibodies. Several genetic polymorphisms in the IL-10 gene have been reported to cause or aggravate certain diseases like inflammatory bowel disease, rheumatoid arthritis, systemic sclerosis, asthma, etc. However, the disease susceptibility and abnormal function of the mutated IL-10 variants remain obscure.
Results
In this study, we used seven bioinformatics tools (SIFT, PROVEAN, PMut, PANTHER, PolyPhen-2, PHD-SNP, and SNPs&GO) to predict the disease susceptible non-synonymous SNPs (nsSNPs) of IL-10. Nine nsSNPs of IL-10 were predicted to be potentially deleterious: R42G, R45Q, F48L, E72G, M95T, A98D, R125S, Y155C, and I168T. Except two, all of the putative deleterious mutations are found in the highly conserved region of IL-10 protein structure, thus affecting the protein's stability. The 3-D structure of mutant proteins was modeled by project HOPE, and the protein–protein interactions were assessed with STRING. The predicted nsSNPs: R42Q, R45Q, F48L, E72G, and I168T are situated in the binding site region of the IL-10R1 receptor. Disruption of binding affinity with its receptor leads to deregulation of the JAK-STAT pathway and results in enhanced inflammation that imbalance in cellular signaling. Finally, Kaplan–Meier Plotter analysis displayed that deregulation of IL-10 expression affects gastric and ovarian cancer patients' survival rate. Thus, IL-10 could be useful as a potential prognostic marker gene for some cancers.
Conclusion
This study has determined the deleterious nsSNPs of IL-10 that might contribute to the malfunction of IL-10 protein and ultimately lead to the IL-10 associated diseases.
Collapse
|
30
|
Palano MT, Gallazzi M, Cucchiara M, Dehò F, Capogrosso P, Bruno A, Mortara L. The tumor innate immune microenvironment in prostate cancer: an overview of soluble factors and cellular effectors. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:694-718. [PMID: 36338516 PMCID: PMC9630328 DOI: 10.37349/etat.2022.00108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/12/2022] [Indexed: 01/14/2023] Open
Abstract
Prostate cancer (PCa) accounts as the most common non-cutaneous disease affecting males, and as the first cancer, for incidence, in male. With the introduction of the concept of immunoscore, PCa has been classified as a cold tumor, thus driving the attention in the development of strategies aimed at blocking the infiltration/activation of immunosuppressive cells, while favoring the infiltration/activation of anti-tumor immune cells. Even if immunotherapy has revolutionized the approaches to cancer therapy, there is still a window failure, due to the immune cell plasticity within PCa, that can acquire pro-tumor features, subsequent to the tumor microenvironment (TME) capability to polarize them. This review discussed selected relevant soluble factors [transforming growth factor-beta (TGFβ), interleukin-6 (IL-6), IL-10, IL-23] and cellular components of the innate immunity, as drivers of tumor progression, immunosuppression, and angiogenesis within the PCa-TME.
Collapse
Affiliation(s)
- Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy
| | - Matteo Gallazzi
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Martina Cucchiara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Federico Dehò
- Unit of Urology, ASST-Sette Laghi, Ospedale di Circolo e Fondazione Macchi, University of Insubria, 21100 Varese, Italy
| | - Paolo Capogrosso
- Unit of Urology, ASST-Sette Laghi, Ospedale di Circolo e Fondazione Macchi, University of Insubria, 21100 Varese, Italy
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy,Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy,Correspondence: Antonino Bruno,
| | - Lorenzo Mortara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy,Lorenzo Mortara, . Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
31
|
Yang E, Chua W, Ng W, Roberts TL. Peripheral Cytokine Levels as a Prognostic Indicator in Gastric Cancer: A Review of Existing Literature. Biomedicines 2021; 9:1916. [PMID: 34944729 PMCID: PMC8698340 DOI: 10.3390/biomedicines9121916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Although strong connections exist between the carcinogenesis of gastric cancer and chronic inflammation, gastric cancer is unique in that the chronic gastritis which frequently precedes carcinogenesis is strongly associated with H. pylori infection. The interplay between H. pylori virulence factors and host immune cells is complex but culminates in the activation of inflammatory pathways and transcription factors such as NF-κB, STAT3, and AP-1, all of which upregulate cytokine production. Due to the key role of cytokines in modulating the immune response against tumour cells as well as possibly stimulating tumour growth and proliferation, different patterns of cytokine secretion may be associated with varying patient outcomes. In relation to gastric cancer, interleukin-6, 8, 10, 17A, TNF, and IFN-γ may have pro-tumour properties, although interleukin-10, TNF, and IFN-γ may have anti-tumour effects. However, due to the lack of studies investigating patient outcomes, only a link between higher interleukin-6 levels and poorer prognosis has been demonstrated. Further investigations which link peripheral cytokine levels to patient prognosis may elucidate important pathological mechanisms in gastric cancer which adversely impact patient survival and allow treatments targeting these processes to be developed.
Collapse
Affiliation(s)
- Elton Yang
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Weng Ng
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| |
Collapse
|
32
|
Mesenchymal-Stromal Cell-like Melanoma-Associated Fibroblasts Increase IL-10 Production by Macrophages in a Cyclooxygenase/Indoleamine 2,3-Dioxygenase-Dependent Manner. Cancers (Basel) 2021; 13:cancers13246173. [PMID: 34944793 PMCID: PMC8699649 DOI: 10.3390/cancers13246173] [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: 09/29/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary Melanoma is the deadliest form of skin cancer, and the number of newly diagnosed cases is on the rise. In recent years, it has become evident that melanoma-associated fibroblasts (MAFs), which surround the melanoma cells, play a key role in tumor growth and its ability to evade immune attack. We found that MAFs resemble bone marrow mesenchymal stromal cells (MSCs), and on the basis of this, we looked for effects that they might have on macrophages. Like MSCs, MAFs cause macrophages to produce IL-10, an anti-inflammatory agent. IL-10 contributes to cancer growth by suppressing natural anti-cancer immunity and can also interfere with anti-melanoma immunotherapies. Our findings may open new avenues for the development of anti-melanoma treatments based on MAF-macrophage interactions. Abstract Melanoma-associated fibroblasts (MAFs) are integral parts of melanoma, providing a protective network for melanoma cells. The phenotypical and functional similarities between MAFs and mesenchymal stromal cells (MSCs) prompted us to investigate if, similarly to MSCs, MAFs are capable of modulating macrophage functions. Using immunohistochemistry, we showed that MAFs and macrophages are in intimate contact within the tumor stroma. We then demonstrated that MAFs indeed are potent inducers of IL-10 production in various macrophage types in vitro, and this process is greatly augmented by the presence of treatment-naïve and chemotherapy-treated melanoma cells. MAFs derived from thick melanomas appear to be more immunosuppressive than those cultured from thin melanomas. The IL-10 increasing effect is mediated, at least in part, by cyclooxygenase and indoleamine 2,3-dioxygenase. Our data indicate that MAF-induced IL-10 production in macrophages may contribute to melanoma aggressiveness, and targeting the cyclooxygenase and indoleamine 2,3-dioxygenase pathways may abolish MAF–macrophage interactions.
Collapse
|
33
|
Jimbu L, Mesaros O, Neaga A, Nanut AM, Tomuleasa C, Dima D, Bocsan C, Zdrenghea M. The Potential Advantage of Targeting Both PD-L1/PD-L2/PD-1 and IL-10-IL-10R Pathways in Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2021; 14:1105. [PMID: 34832887 PMCID: PMC8620891 DOI: 10.3390/ph14111105] [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: 09/10/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 12/30/2022] Open
Abstract
Tumor cells promote the suppression of host anti-tumor type 1 T cell responses by various mechanisms, including the upregulation of surface inhibitory molecules such as programmed death ligand (PD-L)-1, and the production of immunosuppressive cytokines such as interleukin-10 (IL-10). There are over 2000 trials investigating PD-L1 and/or its receptor programmed-death 1 (PD-1) blockade in cancer, leading to the approval of PD-1 or PD-L1 inhibitors in several types of solid cancers and in hematological malignancies. The available data suggest that the molecule PD-L1 on antigen-presenting cells suppresses type 1 T cell immune responses such as cytotoxicity, and that the cytokine IL-10, in addition to downregulating immune responses, increases the expression of inhibitory molecule PD-L1. We hypothesize that the manipulation of both the co-inhibitory network (with anti-PD-L1 blocking antibodies) and suppressor network (with anti-IL-10 blocking antibodies) is an attractive immunotherapeutic intervention for acute myeloid leukemia (AML) patients ineligible for standard treatment with chemotherapy and hematopoietic stem cell transplantation, and with less severe adverse reactions. The proposed combination of these two immunotherapies represents a new approach that can be readily translated into the clinic to improve the therapeutic efficacy of AML disease treatment.
Collapse
Affiliation(s)
- Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, 19-21 Croitorilor Str., 400162 Cluj-Napoca, Romania
| | - Alexandra Neaga
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
| | - Ana Maria Nanut
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Corina Bocsan
- Department of Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania;
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| |
Collapse
|
34
|
Association of Interleukin-10 Polymorphisms with Susceptibility to Colorectal Cancer and Gastric Cancer: an Updated Meta-analysis Based on 106 Studies. J Gastrointest Cancer 2021; 53:1066-1082. [PMID: 34694592 DOI: 10.1007/s12029-021-00685-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND The purpose of this study was to explore the association of IL-10 polymorphisms with susceptibility to colorectal cancer (CRC) and gastric cancer (GC). METHODS PubMed, Scopus, Embase, SciELO, medRxiv, China Biology Medicine Disc, DeepDyve, CNKI, and Web of Science were used to identify all relevant articles published up to 20th June 2021, without any restrictions on ethnicity. Summary odds ratios (ORs) with 95% confidence intervals (CIs) were used to determine the strength of the associations. RESULTS A total of 106 case-control studies were included. For CRC, 15 studies with 2772 cases and 3719 controls on -1082A/G, 11 studies with 3259 cases and 4992 controls on -592C/A, and 3 studies with 477 cases and 544 controls on -819 T/C were selected. For GC, 31 studies with 6229 cases and 8666 controls on -1082A/G, 27 studies with 5457 cases and 8381 controls on -592C/A, and 19 studies with 3556 cases and 6218 controls on -819 T/C were included. Pooled data showed a significant association between IL-10-819 T/C polymorphism and CRC susceptibility in overall population, but not for IL-10-1082A/G and -592C/A polymorphisms. However, IL-10-592C/A polymorphism was associated with CRC risk in Asians. A significant association of IL-10-1082A/G polymorphism with the GC risk was found. In the ethnicity subgroup analysis, a significant association was found between IL-10-1082A/G polymorphism and GC risk among Asians. The IL-10-819 T/C was not associated with GC risk in overall population and by ethnicity. CONCLUSIONS Our pooled data show a significant association of IL-10-819 T/C and IL-10-1082A/G polymorphisms with CRC and GC in overall population, respectively. However, other factors may influence these associations, and large-scale studies with adequate methodological quality are necessary to confirm the impact on CRC and GC risk.
Collapse
|
35
|
Mantle cell lymphoma polarizes tumor-associated macrophages into M2-like macrophages, which in turn promote tumorigenesis. Blood Adv 2021; 5:2863-2878. [PMID: 34297045 DOI: 10.1182/bloodadvances.2020003871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Tumor-associated macrophages (TAMs) are recognized as a hallmark of certain solid cancers and predictors of poor prognosis; however, the functional role of TAMs in lymphoid malignancies, including B-cell lymphoma, has not been well defined. We identified infiltration of F4/80+ TAMs in a syngeneic mouse model using the recently generated murine mantle cell lymphoma (MCL) cell line FC-muMCL1. Multicolor flow cytometric analysis of syngeneic lymphoma tumors showed distinct polarization of F4/80+ TAMs into CD206+ M2 and CD80+ M1 phenotypes. Using human MCL cell lines (Mino, Granta, and JVM2), we further showed that MCL cells polarized monocyte-derived macrophages toward an M2-like phenotype, as assessed by CD163+ expression and increased interleukin-10 (IL-10) level; however, levels of the M1 markers CD80 and IL-12 remained unaffected. To show that macrophages contribute to MCL tumorigenesis, we xenografted the human MCL cell line Mino along with CD14+ monocytes and compared tumor growth between these 2 groups. Results showed that xenografted Mino along with CD14+ monocytes significantly increased the tumor growth in vivo compared with MCL cells alone (P < .001), whereas treatment with liposomal clodronate (to deplete the macrophages) reversed the effect of CD14+ monocytes on growth of MCL xenografts (P < .001). Mechanistically, IL-10 secreted by MCL-polarized M2-like macrophages was found to be responsible for increasing MCL growth by activating STAT1 signaling, whereas IL-10 neutralizing antibody or STAT1 inhibition by fludarabine or STAT1 short hairpin RNA significantly abolished MCL growth (P < .01). Collectively, our data show the existence of a tumor microenvironmental network of macrophages and MCL tumor and suggest the importance of macrophages in interventional therapeutic strategies against MCL and other lymphoid malignancies.
Collapse
|
36
|
Kartikasari AER, Huertas CS, Mitchell A, Plebanski M. Tumor-Induced Inflammatory Cytokines and the Emerging Diagnostic Devices for Cancer Detection and Prognosis. Front Oncol 2021; 11:692142. [PMID: 34307156 PMCID: PMC8294036 DOI: 10.3389/fonc.2021.692142] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic inflammation generated by the tumor microenvironment is known to drive cancer initiation, proliferation, progression, metastasis, and therapeutic resistance. The tumor microenvironment promotes the secretion of diverse cytokines, in different types and stages of cancers. These cytokines may inhibit tumor development but alternatively may contribute to chronic inflammation that supports tumor growth in both autocrine and paracrine manners and have been linked to poor cancer outcomes. Such distinct sets of cytokines from the tumor microenvironment can be detected in the circulation and are thus potentially useful as biomarkers to detect cancers, predict disease outcomes and manage therapeutic choices. Indeed, analyses of circulating cytokines in combination with cancer-specific biomarkers have been proposed to simplify and improve cancer detection and prognosis, especially from minimally-invasive liquid biopsies, such as blood. Additionally, the cytokine signaling signatures of the peripheral immune cells, even from patients with localized tumors, are recently found altered in cancer, and may also prove applicable as cancer biomarkers. Here we review cytokines induced by the tumor microenvironment, their roles in various stages of cancer development, and their potential use in diagnostics and prognostics. We further discuss the established and emerging diagnostic approaches that can be used to detect cancers from liquid biopsies, and additionally the technological advancement required for their use in clinical settings.
Collapse
Affiliation(s)
- Apriliana E. R. Kartikasari
- Translational Immunology and Nanotechnology Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Cesar S. Huertas
- Integrated Photonics and Applications Centre (InPAC), School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Arnan Mitchell
- Integrated Photonics and Applications Centre (InPAC), School of Engineering, RMIT University, Melbourne, VIC, Australia
| | - Magdalena Plebanski
- Translational Immunology and Nanotechnology Research Program, School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
37
|
Targeting STAT3 signaling overcomes gefitinib resistance in non-small cell lung cancer. Cell Death Dis 2021; 12:561. [PMID: 34059647 PMCID: PMC8166856 DOI: 10.1038/s41419-021-03844-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 02/06/2023]
Abstract
Lung cancer is one of the most aggressive cancers with poor prognosis and high resistance rate. The family of signal transducer and activator of transcriptions (STATs) appears to modulate resistance in non-small cell lung cancer (NSCLC). In this work, we demonstrated that STAT3/ZEB1 is a critical axis in gefitinib resistance. STAT3-targeted inhibition therefore is a new potential therapeutic strategy for gefitinib resistance in lung cancer. Our small molecule screening identified a relatively specific STAT3-targeted inhibitor, LL1. Pharmacological and biochemical studies indicated that LL1 block the activation of STAT3 via inhibiting its phosphorylation. Further in vitro and in vivo studies elucidated that LL1 sensitizes the resistance cells to gefitinib through depleting STAT3 activity and blocking STAT3/ZEB1 signaling pathways. Little toxicity of LL1 was observed in animal models. All these favorable results indicated that LL1 is a chemotherapeutic adjuvant for gefitinib resistance in NSCLC.
Collapse
|
38
|
Maimon A, Levi-Yahid V, Ben-Meir K, Halpern A, Talmi Z, Priya S, Mizraji G, Mistriel-Zerbib S, Berger M, Baniyash M, Loges S, Burstyn-Cohen T. Myeloid cell-derived PROS1 inhibits tumor metastasis by regulating inflammatory and immune responses via IL-10. J Clin Invest 2021; 131:126089. [PMID: 33848267 DOI: 10.1172/jci126089] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Stimulation of TAM (TYRO3, AXL, and MERTK) receptor tyrosine kinases promotes tumor progression through numerous cellular mechanisms. TAM cognate ligands GAS6 and PROS1 (for TYRO3 and MERTK) are secreted by host immune cells, an interaction which may support tumor progression. Here, we revealed an unexpected antimetastatic role for myeloid-derived PROS1: suppressing metastatic potential in lung and breast tumor models. Pros1 deletion in myeloid cells led to increased lung metastasis, independent of primary tumor infiltration. PROS1-cKO bone marrow-derived macrophages (BMDMs) led to elevated TNF-α, IL-6, Nos2, and IL-10 via modulation of the Socs3/NF-κB pathway. Conditioned medium from cKO BMDMs enhanced EMT, ERK, AKT, and STAT3 activation within tumor cells and promoted IL-10-dependent invasion and survival. Macrophages isolated from metastatic lungs modulated T cell proliferation and function, as well as expression of costimulatory molecules on DCs in a PROS1-dependent manner. Inhibition of MERTK kinase activity blocked PROS1-mediated suppression of TNF-α and IL-6 but not IL-10. Overall, using lung and breast cancer models, we identified the PROS1/MERTK axis within BMDMs as a potent regulator of adaptive immune responses with a potential to suppress metastatic seeding and revealed IL-10 regulation by PROS1 to deviate from that of TNF-α and IL-6.
Collapse
Affiliation(s)
- Avi Maimon
- The Institute for Dental Sciences, Faculty of Dental Medicine and
| | | | - Kerem Ben-Meir
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, the Hebrew University, Jerusalem, Israel
| | - Amit Halpern
- The Institute for Dental Sciences, Faculty of Dental Medicine and
| | - Ziv Talmi
- The Institute for Dental Sciences, Faculty of Dental Medicine and
| | - Shivam Priya
- The Institute for Dental Sciences, Faculty of Dental Medicine and
| | - Gabriel Mizraji
- The Institute for Dental Sciences, Faculty of Dental Medicine and
| | - Shani Mistriel-Zerbib
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, the Hebrew University, Jerusalem, Israel
| | - Michael Berger
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, the Hebrew University, Jerusalem, Israel
| | - Michal Baniyash
- The Lautenberg Center for Immunology and Cancer Research, Israel-Canada Medical Research Institute, Faculty of Medicine, the Hebrew University, Jerusalem, Israel
| | - Sonja Loges
- Division of Personalized Medical Oncology (A420), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Personalized Oncology, University Hospital Mannheim, Mannheim, Germany
| | | |
Collapse
|
39
|
Mantilla-Rojas C, Yu M, Rinella ES, Lynch RM, Perry A, Jaimes-Alvarado J, Anderson KR, Barba E, Bourgeois EJ, Konganti K, Threadgill DW. A molecular subtype of colorectal cancers initiates independently of epidermal growth factor receptor and has an accelerated growth rate mediated by IL10-dependent anergy. Oncogene 2021; 40:3047-3059. [PMID: 33767440 PMCID: PMC9113393 DOI: 10.1038/s41388-021-01752-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/28/2021] [Accepted: 03/08/2021] [Indexed: 02/01/2023]
Abstract
Although epidermal growth factor receptor (EGFR)-targeted therapies are approved for colorectal cancer (CRC) treatment, only 15% of CRC patients respond to EGFR inhibition. Here, we show that colorectal cancers (CRC) can initiate and grow faster through an EGFR-independent mechanism, irrespective of the presence of EGFR, in two different mouse models using tissue-specific ablation of Egfr. The growth benefit in the absence of EGFR is also independent of Kras status. An EGFR-independent gene expression signature, also observed in human CRCs, revealed that anergy-inducing genes are overexpressed in EGFR-independent polyps, suggesting increased infiltration of anergic lymphocytes promotes an accelerated growth rate that is partially caused by escape from cell-mediated immune responses. Many genes in the EGFR-independent gene expression signature are downstream targets of interleukin 10 receptor alpha (IL10RA). We further show that IL10 is detectable in serum from mice with EGFR-independent colon polyps. Using organoids in vitro and Src ablation in vivo, we show that IL10 contributes to growth of EGFR-independent CRCs, potentially mediated by the well-documented role of SRC in IL10 signaling. Based on these data, we show that the combination of an EGFR inhibitor with an anti-IL10 neutralizing antibody results in decreased cell proliferation in organoids and in decreased polyp size in pre-clinical models harboring EGFR-independent CRCs, providing a new therapeutic intervention for CRCs resistant to EGFR inhibitor therapies.
Collapse
Affiliation(s)
- Carolina Mantilla-Rojas
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
| | - Ming Yu
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Erica S Rinella
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- GeneDx, Gaithersburg, MD, USA
| | - Rachel M Lynch
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Amie Perry
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
- Thompson Bishop Sparks State Diagnostic Laboratory, Auburn, AL, USA
| | - Jorge Jaimes-Alvarado
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
| | - Kathryn R Anderson
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
| | - Estefania Barba
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
| | - Evann J Bourgeois
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA
| | - Kranti Konganti
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, USA
| | - David W Threadgill
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA.
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, TX, USA.
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, USA.
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
40
|
PIWIL1 governs the crosstalk of cancer cell metabolism and immunosuppressive microenvironment in hepatocellular carcinoma. Signal Transduct Target Ther 2021; 6:86. [PMID: 33633112 PMCID: PMC7907082 DOI: 10.1038/s41392-021-00485-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 09/24/2020] [Accepted: 10/09/2020] [Indexed: 02/06/2023] Open
Abstract
Altered energy metabolism of cancer cells shapes the immune cell response in the tumor microenvironment that facilitates tumor progression. Herein, we reported the novel of tumor cell-expressed Piwi Like RNA-Mediated Gene Silencing 1 (PIWIL1) in mediating the crosstalk of fatty acid metabolism and immune response of human hepatocellular carcinoma (HCC). PIWIL1 expression in HCC was increased compared to normal hepatic tissues and was positively correlated with the proliferation rate of HCC cell lines. PIWIL1 overexpression accelerated in vitro proliferation and in vivo growth of HCC tumors, while PIWIL1 knockdown showed opposite effects. PIWIL1 increased oxygen consumption and energy production via fatty acid metabolism without altering aerobic glycolysis. Inhibition of fatty acid metabolism abolished PIWIL1-induced HCC proliferation and growth. RNA-seq analysis revealed that immune system regulation might be involved, which was echoed by the experimental observation that PIWIL1-overexpressing HCC cells attracted myeloid-derived suppressor cells (MDSCs) into the tumor microenvironment. MDSCs depletion reduced the proliferation and growth of PIWIL1-overexpressing HCC tumors. Complement C3, whose secretion was induced by PIWIL1 in HCC cells, mediates the interaction of HCC cells with MDSCs by activated p38 MAPK signaling in MDSCs, which in turn initiated expression of immunosuppressive cytokine IL10. Neutralizing IL10 secretion reduced the immunosuppressive activity of MDSCs in the microenvironment of PIWIL1-overexpressing HCC. Taken together, our study unraveled the critical role of PIWIL1 in initiating the interaction of cancer cell metabolism and immune cell response in HCC. Tumor cells-expressed PIWIL1 may be a potential target for the development of novel HCC treatment.
Collapse
|
41
|
Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells 2021; 10:cells10010067. [PMID: 33406733 PMCID: PMC7824562 DOI: 10.3390/cells10010067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/09/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Gastrointestinal cancer is highly associated with inflammatory processes inducing the release of cytokines from cancer or immune cells, including interferons, interleukins, chemokines, colony-stimulating factors, and growth factors, which promote or suppress tumor progression. Inflammatory cytokines within the tumor microenvironment promote immune cell infiltration. Infiltrating immune, and tumor-surrounding stromal cells support tumor growth, angiogenesis, metastasis, and immunosuppression through communication with inflammatory cytokines and cell adhesion molecules. Notably, infiltrating immune and tumor cells present immunosuppressive molecules, such as programmed death-ligand 1 (PD-L1) and CD80/CD86. Suppression of cytotoxic T cells promotes tumor avoidance of immune surveillance and greater malignancy. Moreover, glycosylation and sialylation of proteins hyperexpressed on the cancer cell surface have been shown to enhance immune escape and metastasis. Cytokine treatments and immune checkpoint inhibitors are widely used in clinical practice. However, the tumor microenvironment is a rapidly changing milieu involving several factors. In this review, we have provided a summary of the interactions of inflammation and cell adhesion molecules between cancer and other cell types, to improve understanding of the tumor microenvironment.
Collapse
|
42
|
Mesaros O, Jimbu L, Neaga A, Popescu C, Berceanu I, Tomuleasa C, Fetica B, Zdrenghea M. Macrophage Polarization in Chronic Lymphocytic Leukemia: Nurse-Like Cells Are the Caretakers of Leukemic Cells. Biomedicines 2020; 8:E516. [PMID: 33228048 PMCID: PMC7699370 DOI: 10.3390/biomedicines8110516] [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: 09/14/2020] [Revised: 11/09/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophages are terminally differentiated innate immune cells. Through their activation, they can be polarized towards the pro-inflammatory M1 type or the wound healing-associated, anti-inflammatory M2 type macrophages. In the tumor microenvironment (TME), M2 is the dominant phenotype and these cells are referred to as tumor-associated macrophages (TAMs). TAMs secrete cytokines and chemokines, exerting an antiapoptotic, proliferative and pro-metastatic effect on the tumor cells. TAMs can be found in many cancers, including chronic lymphocytic leukemia (CLL), where they are called nurse-like cells (NLCs). Despite the generally indolent behavior of CLL, the proportion of treatment-refractory patients is significant. As with the majority of cancers, despite significant recent progress, CLL pathogenesis is poorly understood. The emerging role of the TME in nurturing the neoplastic process warrants the investigation of macrophages as a significant pathogenetic element of tumors. In this paper, we review the current knowledge on the role of stromal macrophages in CLL.
Collapse
Affiliation(s)
- Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Alexandra Neaga
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
| | - Cristian Popescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Infectious Diseases, County Emergency Hospital Alba Iulia, 20 Decebal str., 510093 Alba-Iulia, Romania
| | - Iulia Berceanu
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Bogdan Fetica
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes str., 400012 Cluj-Napoca, Romania; (L.J.); (A.N.); (C.P.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Street, 400015 Cluj-Napoca, Romania; (I.B.); (B.F.)
| |
Collapse
|
43
|
Algood HMS. T Cell Cytokines Impact Epithelial Cell Responses during Helicobacter pylori Infection. THE JOURNAL OF IMMUNOLOGY 2020; 204:1421-1428. [PMID: 32152211 DOI: 10.4049/jimmunol.1901307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/08/2019] [Indexed: 12/24/2022]
Abstract
The goal of this Brief Review is to highlight literature that demonstrates how cytokines made by T lymphocytes impact the gastric epithelium, especially during Helicobacter pylori infection. These cytokines effect many of the diverse functions of the epithelium and the epithelium's interactions with H. pylori The focal point of this Brief Review will be on how T cell cytokines impact antimicrobial function and barrier function and how T cell cytokines influence the development and progression of cancer. Furthermore, the modulation of epithelial-derived chemokines by H. pylori infection will be discussed.
Collapse
Affiliation(s)
- Holly M Scott Algood
- Veterans Affairs Tennessee Valley Healthcare Services, Nashville, TN 37212; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37212; and Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37212
| |
Collapse
|
44
|
Wu C, Zeng MH, Liao G, Qian K, Li H. Neuropilin-1 Interacts with Fibronectin-1 to Promote Epithelial-Mesenchymal Transition Progress in Gastric Cancer. Onco Targets Ther 2020; 13:10677-10687. [PMID: 33116644 PMCID: PMC7585825 DOI: 10.2147/ott.s275327] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Neuropilin-1 (NRP1) binds to many ligands and co-receptors and affects cell survival and migration, which is essential for tumor progression. However, there are still largely unknowns about how NRP1 affects the epithelial-mesenchymal transition (EMT)-related malignant progression in gastric cancer. Methods We used TCGA to analyze the expression of NRP1 in gastric cancer and its impact on patient survival. In in vitro experiments, transwell, wound healing and colony formation assays were used to evaluate the effects of NRP1 and ginsenoside Rg3 on the invasion, migration and proliferation of gastric cancer cells. In in vivo experiments, we evaluated the overexpression and knockdown of NRP1 and the effect of ginsenoside Rg3 on tumor growth. Results We found that NRP1 is highly expressed in advanced gastric cancer and associated with poor prognosis. Knockdown of NRP1 expression can inhibit the proliferation and metastasis of gastric cancer cells. Mechanically. NRP1 interacts with fibronectin-1 (FN1) to promote the malignant progression of gastric cancer cells through ECM remodeling. In addition, we found that ginsenoside Rg3 can block the interaction of NRP1 and FN1 and inhibit the progression of gastric cancer. Conclusion Our study suggested that the interaction of NRP1 and FN1 is crucial for the malignant progression of gastric cancer. This may provide a new perspective and potential treatment methods for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Chao Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Meng-Hua Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Gang Liao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Kun Qian
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, People's Republic of China
| |
Collapse
|
45
|
Wang Y, Hu Y, Pan K, Li H, Shang S, Wang Y, Tang G, Han X. In-vivo imaging revealed antigen-directed gingival B10 infiltration in experimental periodontitis. Biochim Biophys Acta Mol Basis Dis 2020; 1867:165991. [PMID: 33080346 DOI: 10.1016/j.bbadis.2020.165991] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 12/15/2022]
Abstract
Our previous study demonstrated that IL-10 secreting B (B10) cells alleviate inflammation and bone loss in experimental periodontitis. The purpose of this study is to determine whether antigen-specificity is required for the local infiltration of B10 cells. Experimental periodontitis was induced in the recipient mice by placement of silk ligature with or without the presence of live Porphyromonas gingivalis (P. gingivalis). Donor mice were pre-immunized by intraperitoneal (IP) injection of formalin-fixed P. gingivalis, or PBS as non-immunized control. Spleen B cells were purified and treated with LPS and CpG for 48 h to expand the B10 population in vitro. Fluorescence-labelled B10 cells were transferred into the recipient mice by tail vein injection and were tracked on day 0, 3, 5 and 10 using IVIS Spectrum in vivo imaging system. The number of B10 cells and P. gingivalis-binding B cells were significantly increased after in vitro treatment of LPS and CpG. On day 5, the fluorescence intensity in gingival tissues was the highest in mice transferred with B10 cells from pre-immunized donor mice. Gingival expression of IL-6, TNF-α, RANKL/OPG ratio and periodontal bone loss in recipient mice were significantly reduced, and the expression of IL-10 and the number of CD19+ B cells were significantly increased after pre-immunized B10 cell transfer in the presence of antigen, compared to those with non-immunized B10 cell transfer or no antigen presence. This study suggests that antigen specificity dictate the local infiltration of B10 cells into periodontal tissue and these antigen-specific B10 cells promote anti-inflammatory responses.
Collapse
Affiliation(s)
- Yufeng Wang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China; Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States
| | - Yang Hu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States
| | - Keqing Pan
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Stomatology, the affiliated hospital of Qingdao University, Qingdao, Shandong 266003, China
| | - Hao Li
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Prosthodontics, the Affiliated Hospital of Stomatology, Guangxi Medical University, Nanning 530021, China
| | - Shu Shang
- Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Yuhua Wang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China
| | - Guoyao Tang
- Department of Oral Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai 200011, China
| | - Xiaozhe Han
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Harvard School of Dental Medicine Affiliate, Cambridge, MA 02142, United States; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, United States.
| |
Collapse
|
46
|
Li J, Liu Y, Liu Q. [Expression of superoxide dismutase 2 in breast cancer and its clinical significance]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:1103-1111. [PMID: 32895185 DOI: 10.12122/j.issn.1673-4254.2020.08.06] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To evaluate the expression and prognostic value of superoxide dismutase 2 (SOD2) in breast cancer and explore its possible role in the occurrence and progression of breast cancer. METHODS We performed bioinformatics analysis of the TCGA data for the expression and clinical relevance of SOD2 in patients with breast cancer. Gene enrichment analysis (GSEA) was performed using the KEGG gene set, the protein interaction network was constructed using the STRING database, and the key genes were screened using Cytoscape software. We also collected 60 pairs of primary breast cancer tissue samples and adjacent samples for detecting SOD2 expressions using immunohistochemistry and RT-qPCR and analyzed the correlation of SOD2 expression with the clinicopathological parameters of the patients. RESULTS The expression of SOD2 was significantly lower in breast cancer tissue than in adjacent tissues with significant correlation with TNM stage and axillary lymph node metastasis (P < 0.05). Kaplan-Meier survival analysis showed that the recurrence-free survival, distant metastasis-free survival (RFS) and post-progressive survival were significantly shorted in patients with high SOD2 expression than in those with low SOD2 expression (P < 0.05). GSEA enrichment analysis indicated that SOD2 played an important role in the JAK-STAT signaling pathway. IL10 and STAT4 were identified as the key genes in the PPI network, and they were both positively correlated with SOD2. In the 60 pairs of clinical samples, SOD2 was highly expressed in breast cancer tissues with close correlation with axillary lymph node metastasis and the expressions of estrogen receptor and androgen receptor (P < 0.05). CONCLUSIONS The expression of SOD2 in breast cancer is significantly correlated with TNM stage and axillary lymph node metastasis. SOD2 may affect the proliferation, invasion and metastasis of breast cancer cells possibly by regulating IL10 and/or STAT4 to affect the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Jinping Li
- General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Yaobang Liu
- General Hospital of Ningxia Medical University, Yinchuan 750004, China
| | - Qilun Liu
- General Hospital of Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
47
|
Prognostic and Predictive Value of Cadherin 11 for Patients with Gastric Cancer and Its Correlation with Tumor Microenvironment: Results from Microarray Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8107478. [PMID: 32685527 PMCID: PMC7335407 DOI: 10.1155/2020/8107478] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/03/2020] [Indexed: 12/29/2022]
Abstract
Gastric cancer is a disease characterized by inflammation, and epithelial-to-mesenchymal transition (EMT) and tumor-associated macrophages (TAMs) both play a vital role in epithelial-driven malignancy. In the present study, we performed an integrated bioinformatics analysis of transcriptome data from multiple databases of gastric cancer patients and worked on a biomarker for evaluating tumor prognosis. We found that cadherin 11 (CDH11) is highly expressed not only in gastric cancer tissues but also in EMT molecular subtypes and metastatic patients. Also, we obtained evidence that CDH11 has a significant correlation with infiltrating immune cells in the tumor microenvironment (TME). Our findings reflected that CDH11 likely plays an important role in tumor immune escape and could provide a prognostic biomarker and potential therapeutic target for patients with gastric cancer.
Collapse
|
48
|
STAT3 Pathway in Gastric Cancer: Signaling, Therapeutic Targeting and Future Prospects. BIOLOGY 2020; 9:biology9060126. [PMID: 32545648 PMCID: PMC7345582 DOI: 10.3390/biology9060126] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Molecular signaling pathways play a significant role in the regulation of biological mechanisms, and their abnormal expression can provide the conditions for cancer development. The signal transducer and activator of transcription 3 (STAT3) is a key member of the STAT proteins and its oncogene role in cancer has been shown. STAT3 is able to promote the proliferation and invasion of cancer cells and induces chemoresistance. Different downstream targets of STAT3 have been identified in cancer and it has also been shown that microRNA (miR), long non-coding RNA (lncRNA) and other molecular pathways are able to function as upstream mediators of STAT3 in cancer. In the present review, we focus on the role and regulation of STAT3 in gastric cancer (GC). miRs and lncRNAs are considered as potential upstream mediators of STAT3 and they are able to affect STAT3 expression in exerting their oncogene or onco-suppressor role in GC cells. Anti-tumor compounds suppress the STAT3 signaling pathway to restrict the proliferation and malignant behavior of GC cells. Other molecular pathways, such as sirtuin, stathmin and so on, can act as upstream mediators of STAT3 in GC. Notably, the components of the tumor microenvironment that are capable of targeting STAT3 in GC, such as fibroblasts and macrophages, are discussed in this review. Finally, we demonstrate that STAT3 can target oncogene factors to enhance the proliferation and metastasis of GC cells.
Collapse
|
49
|
Baj J, Brzozowska K, Forma A, Maani A, Sitarz E, Portincasa P. Immunological Aspects of the Tumor Microenvironment and Epithelial-Mesenchymal Transition in Gastric Carcinogenesis. Int J Mol Sci 2020; 21:E2544. [PMID: 32268527 PMCID: PMC7177728 DOI: 10.3390/ijms21072544] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 03/30/2020] [Accepted: 04/02/2020] [Indexed: 12/11/2022] Open
Abstract
Infection with Helicobacter pylori, a Gram-negative, microaerophilic pathogen often results in gastric cancer in a subset of affected individuals. This explains why H. pylori is the only bacterium classified as a class I carcinogen by the World Health Organization. Several studies have pinpointed mechanisms by which H. pylori alters signaling pathways in the host cell to cause diseases. In this article, the authors have reviewed 234 studies conducted over a span of 18 years (2002-2020). The studies investigated the various mechanisms associated with gastric cancer induction. For the past 1.5 years, researchers have discovered new mechanisms contributing to gastric cancer linked to H. pylori etiology. Alongside alteration of the host signaling pathways using oncogenic CagA pathways, H. pylori induce DNA damage in the host and alter the methylation of DNA as a means of perturbing downstream signaling. Also, with H. pylori, several pathways in the host cell are activated, resulting in epithelial-to-mesenchymal transition (EMT), together with the induction of cell proliferation and survival. Studies have shown that H. pylori enhances gastric carcinogenesis via a multifactorial approach. What is intriguing is that most of the targeted mechanisms and pathways appear common with various forms of cancer.
Collapse
Affiliation(s)
- Jacek Baj
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (A.M.)
| | - Karolina Brzozowska
- Chair and Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Alicja Forma
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (A.M.)
| | - Amr Maani
- Chair and Department of Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (A.F.); (A.M.)
| | - Elżbieta Sitarz
- Chair and 1st Department of Psychiatry, Psychotherapy and Early Intervention, Medical University of Lublin, Gluska Street 1, 20-439 Lublin, Poland;
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, 70124 Bari, Italy;
| |
Collapse
|
50
|
Sarode P, Schaefer MB, Grimminger F, Seeger W, Savai R. Macrophage and Tumor Cell Cross-Talk Is Fundamental for Lung Tumor Progression: We Need to Talk. Front Oncol 2020; 10:324. [PMID: 32219066 PMCID: PMC7078651 DOI: 10.3389/fonc.2020.00324] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 02/24/2020] [Indexed: 12/14/2022] Open
Abstract
Regardless of the promising results of certain immune checkpoint blockers, current immunotherapeutics have met a bottleneck concerning response rate, toxicity, and resistance in lung cancer patients. Accumulating evidence forecasts that the crosstalk between tumor and immune cells takes center stage in cancer development by modulating tumor malignancy, immune cell infiltration, and immune evasion in the tumor microenvironment (TME). Cytokines and chemokines secreted by this crosstalk play a major role in cancer development, progression, and therapeutic management. An increased infiltration of Tumor-associated macrophages (TAMs) was observed in most of the human cancers, including lung cancer. In this review, we emphasize the role of cytokines and chemokines in TAM-tumor cell crosstalk in the lung TME. Given the role of cytokines and chemokines in immunomodulation, we propose that TAM-derived cytokines and chemokines govern the cancer-promoting immune responses in the TME and offer a new immunotherapeutic option for lung cancer treatment.
Collapse
Affiliation(s)
- Poonam Sarode
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Martina Barbara Schaefer
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany
| | - Rajkumar Savai
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany.,Department of Internal Medicine, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Justus Liebig University, Giessen, Germany.,Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|