1
|
Chun KS, Kim EH, Kim DH, Song NY, Kim W, Na HK, Surh YJ. Targeting cyclooxygenase-2 for chemoprevention of inflammation-associated intestinal carcinogenesis: An update. Biochem Pharmacol 2024; 228:116259. [PMID: 38705538 DOI: 10.1016/j.bcp.2024.116259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Mounting evidence from preclinical and clinical studies suggests that persistent inflammation functions as a driving force in the journey to cancer. Cyclooxygenase-2 (COX-2) is a key enzyme involved in inflammatory signaling. While being transiently upregulated upon inflammatory stimuli, COX-2 has been found to be consistently overexpressed in human colorectal cancer and several other malignancies. The association between chronic inflammation and cancer has been revisited: cancer can arise when inflammation fails to resolve. Besides its proinflammatory functions, COX-2 also catalyzes the production of pro-resolving as well as anti-inflammatory metabolites from polyunsaturated fatty acids. This may account for the side effects caused by long term use of some COX-2 inhibitory drugs during the cancer chemopreventive trials. This review summarizes the latest findings highlighting the dual functions of COX-2 in the context of its implications in the development, maintenance, and progression of cancer.
Collapse
Affiliation(s)
- Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Korea
| | - Eun-Hee Kim
- College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Seongnam 13488, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Na-Young Song
- Department of Oral Biology, BK21 Four Project, Yonsei University College of Dentistry, Seoul 03722, South Korea
| | - Wonki Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea
| | - Hye-Kyung Na
- Department of Food Science and Biotechnology, College of Knowledge-Based Services Engineering, Sungshin Women's University, Seoul 01133, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea.
| |
Collapse
|
2
|
Kim NH, Lee J, Kim SH, Kang SH, Bae S, Yu CH, Seo J, Kim HT. ALK5/VEGFR2 dual inhibitor TU2218 alone or in combination with immune checkpoint inhibitors enhances immune-mediated antitumor effects. Cancer Immunol Immunother 2024; 73:190. [PMID: 39105882 PMCID: PMC11303640 DOI: 10.1007/s00262-024-03777-4] [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: 04/04/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024]
Abstract
Transforming growth factor β (TGFβ) is present in blood of patients who do not respond to anti-programmed cell death (ligand) 1 [PD-(L)1] treatment, and through synergy with vascular endothelial growth factor (VEGF), it helps to create an environment that promotes tumor immune evasion and immune tolerance. Therefore, simultaneous inhibition of TGFβ/VEGF is more effective than targeting TGFβ alone. In this study, the dual inhibitory mechanism of TU2218 was identified through in vitro analysis mimicking the tumor microenvironment, and its antitumor effects were analyzed using mouse syngeneic tumor models. TU2218 directly restored the activity of damaged cytotoxic T lymphocytes (CTLs) and natural killer cells inhibited by TGFβ and suppressed the activity and viability of regulatory T cells. The inactivation of endothelial cells induced by VEGF stimulation was completely ameliorated by TU2218, an effect not observed with vactosertib, which inhibits only TGFβ signaling. The combination of TU2218 and anti-PD1 therapy had a significantly greater antitumor effect than either drug alone in the poorly immunogenic B16F10 syngeneic tumor model. The mechanism of tumor reduction was confirmed by flow cytometry, which showed upregulated VCAM-1 expression in vascular cells and increased influx of CD8 + CTLs into the tumor. As another strategy, combination of anti-CTLA4 therapy and TU2218 resulted in high complete regression (CR) rates in CT26 and WEHI-164 tumor models. In particular, immunological memory generated by the combination of anti-CTLA4 and TU2218 in the CT26 model prevented the development of tumors after additional tumor cell transplantation, suggesting that the TU2218-based combination has therapeutic potential in immunotherapy.
Collapse
Affiliation(s)
- Nam-Hoon Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jihyun Lee
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seung-Hyun Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Seong-Ho Kang
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Sowon Bae
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chan-Hee Yu
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jeongmin Seo
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Hun-Taek Kim
- TiumBio Co., Ltd. Seongnam-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
3
|
Bakrim S, El Hachlafi N, Khalid A, Abdalla AN, El Omari N, Aboulaghras S, Sakran AM, Goh KW, Ming LC, Razi P, Bouyahya A. Recent advances and molecular mechanisms of TGF-β signaling in colorectal cancer, with focus on bioactive compounds targeting. Biomed Pharmacother 2024; 177:116886. [PMID: 38945700 DOI: 10.1016/j.biopha.2024.116886] [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: 03/03/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most significant forms of human cancer. It is characterized by its heterogeneity because several molecular factors are involved in contiguity and can link it to others without having a linear correlation. Among the factors influencing tumor transformation in CRC, transforming growth factor-beta (TGF-β) plays a key promoter role. This factor is associated with human colorectal tumors with a very high prognosis: it increases the survival, invasion, and metastasis of CRC cells, thus functioning as an oncogene. The inhibition of this factor can constitute a major therapeutic route for CRC treatment. Various chemical drugs including synthetic molecules and biotherapies have been developed as TGF-β inhibitors. Moreover, the scientific community has recently shown a major interest in screening natural drugs inhibiting TGF-β in CRC. In this context, we carried out this review article using computerized databases, such as PubMed, Google Scholar, Springer Link, Science Direct, Cochrane Library, Embase, Web of Science, and Scopus, to highlight the molecular mechanism of TGF-β in CRC induction and progression and current advances in the pharmacodynamic effects of natural bioactive substances targeting TGF-β in CRC.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir 80000, Morocco
| | - Naoufal El Hachlafi
- Microbial Biotechnology and Bioactive Molecules Laboratory, Sciences and Technologies Faculty, Sidi Mohmed Ben Abdellah University, P.O.Box-2002, Imouzzer Road, Fez, Morocco
| | - Asaad Khalid
- Health Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Sara Aboulaghras
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| | - Ashraf M Sakran
- Department of Anatomy, Faculty of Medicine, Umm Alqura University, Makkah 21955, Saudi Arabia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai, Malaysia
| | - Long Chiau Ming
- School of Medical and Life Sciences, Sunway University, Sunway City, Malaysia.
| | - Pakhrur Razi
- Center of Disaster Monitoring and Earth Observation, Universitas Negeri Padang, Padang, Indonesia.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat 10106, Morocco
| |
Collapse
|
4
|
Khan A, Zhang Y, Ma N, Shi J, Hou Y. NF-κB role on tumor proliferation, migration, invasion and immune escape. Cancer Gene Ther 2024:10.1038/s41417-024-00811-6. [PMID: 39033218 DOI: 10.1038/s41417-024-00811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Nuclear factor kappa-B (NF-κB) is a nuclear transcription factor that plays a key factor in promoting inflammation, which can lead to the development of cancer in a long-lasting inflammatory environment. The activation of NF-κB is essential in the initial phases of tumor development and progression, occurring in both pre-malignant cells and cells in the microenvironment such as phagocytes, T cells, and B cells. In addition to stimulating angiogenesis, inhibiting apoptosis, and promoting the growth of tumor cells, NF-κB activation also causes the epithelial-mesenchymal transition, and tumor immune evasion. Therapeutic strategies that focus on immune checkpoint molecules have revolutionized cancer treatment by enabling the immune system to activate immunological responses against tumor cells. This review focused on understanding the NF-κB signaling pathway in the context of cancer.
Collapse
Affiliation(s)
- Afrasyab Khan
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, The People's Republic of China
| | - Yao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, The People's Republic of China
| | - Ningna Ma
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, The People's Republic of China
| | - Juanjuan Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, The People's Republic of China
| | - Yongzhong Hou
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu Province, The People's Republic of China.
| |
Collapse
|
5
|
Xu S, Deng KQ, Lu C, Fu X, Zhu Q, Wan S, Zhang L, Huang Y, Nie L, Cai H, Wang Q, Zeng H, Zhang Y, Wang F, Ren H, Chen Y, Yan H, Xu K, Zhou L, Lu M, Zhu Y, Liu S, Lu Z. Interleukin-6 classic and trans-signaling utilize glucose metabolism reprogramming to achieve anti- or pro-inflammatory effects. Metabolism 2024; 155:155832. [PMID: 38438106 DOI: 10.1016/j.metabol.2024.155832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/26/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
Interleukin (IL)-6 has anti- and pro-inflammatory functions, controlled by IL-6 classic and trans-signaling, respectively. Differences in the downstream signaling mechanism between IL-6 classic and trans-signaling have not been identified. Here, we report that IL-6 activates glycolysis to regulate the inflammatory response. IL-6 regulates glucose metabolism by forming a complex containing signal-transducing activators of transcription 3 (STAT3), hexokinase 2 (HK2), and voltage-dependent anion channel 1 (VDAC1). The IL-6 classic signaling directs glucose flux to oxidative phosphorylation (OxPhos), while IL-6 trans-signaling directs glucose flux to anaerobic glycolysis. Classic IL-6 signaling promotes STAT3 translocation into mitochondria to interact with pyruvate dehydrogenase kinase-1 (PDK1), leading to pyruvate dehydrogenase α (PDHA) dissociation from PDK1. As a result, PDHA is dephosphorylated, and STAT3 is phosphorylated at Ser727. By contrast, IL-6 trans-signaling promotes the interaction of sirtuin 2 (SIRT2) and lactate dehydrogenase A (LDHA), leading to the dissociation of STAT3 from SIRT2. As a result, LDHA is deacetylated, and STAT3 is acetylated and phosphorylated at Tyr705. IL-6 classic signaling promotes the differentiation of regulatory T cells via the PDK1/STAT3/PDHA axis, whereas IL-6 trans-signaling promotes the differentiation of Th17 cells via the SIRT2/STAT3/LDHA axis. Conclusion: IL-6 classic signaling generates anti-inflammatory functions by shifting energy metabolism to OxPhos, while IL-6 trans-signaling generates pro-inflammatory functions by shifting energy metabolism to anaerobic glycolysis.
Collapse
Affiliation(s)
- Shilei Xu
- Department of General Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510530, China.
| | - Ke-Qiong Deng
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| | - Chengbo Lu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| | - Xin Fu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China
| | - Qingmei Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Shiqi Wan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Lin Zhang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China
| | - Yu Huang
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China.
| | - Longyu Nie
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China.
| | - Huanhuan Cai
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| | - Qiming Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Human Province, China
| | - Hao Zeng
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, China.
| | - Yufeng Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, China.
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China
| | - Hong Ren
- Shanghai Children's Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, China.
| | - Yu Chen
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Huan Yan
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Li Zhou
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen 45122, Germany.
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China; State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Taikang Center for Life and Medical Sciences, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430072, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Human Province, China.
| | - Zhibing Lu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
6
|
Wang Z, Chang Y, Sun H, Li Y, Tang T. Advances in molecular mechanisms of inflammatory bowel disease‑associated colorectal cancer (Review). Oncol Lett 2024; 27:257. [PMID: 38646499 PMCID: PMC11027113 DOI: 10.3892/ol.2024.14390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/15/2024] [Indexed: 04/23/2024] Open
Abstract
The link between inflammation and cancer is well documented and colonic inflammation caused by inflammatory bowel disease (IBD) is thought to be a high-risk factor for the development of colorectal cancer (CRC). The complex crosstalk between epithelial and inflammatory cells is thought to underlie the progression from inflammation to cancer. The present review collates and summarises recent advances in the understanding of the pathogenesis of IBD-associated CRC (IBD-CRC), including the oncogenic mechanisms of the main inflammatory signalling pathways and genetic alterations induced by oxidative stress during colonic inflammation, and discusses the crosstalk between the tumour microenvironment, intestinal flora and host immune factors during inflammatory oncogenesis in colitis-associated CRC. In addition, the therapeutic implications of anti-inflammatory therapy for IBD-CRC were discussed, intending to provide new insight into improve clinical practice.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yu Chang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Haibo Sun
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yuqin Li
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Tongyu Tang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
7
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [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: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
8
|
Gulubova M, Chonov D, Aleksandrova E, Ivanova K, Ignatova MM, Vlaykova T. Interleukin-6-Positive Immune Cells as a Possible New Immunologic Marker Associated With the Colorectal Cancer Prognosis. Appl Immunohistochem Mol Morphol 2024; 32:233-243. [PMID: 38712586 PMCID: PMC11073565 DOI: 10.1097/pai.0000000000001198] [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: 06/02/2023] [Accepted: 03/15/2024] [Indexed: 05/08/2024]
Abstract
Chronic inflammation creates tumor microenvironment (TME) that facilitates colorectal cancer (CRC) cell proliferation, migration, metastasis, and tumor progression. Interleukin-6 (IL-6) is a proinflammatory cytokine with a pleiotropic effect on CRC development. We aimed to evaluate IL-6 expression in tumor cells and in immune cells in TME, to assess the serum level and IL6 -174 G/C genotype distribution and to correlate the results with selected morphologic and clinical parameters that may add useful information in understanding the mechanisms of human CRC progression. A total of 153 patients with CRC were recruited in the current study. We assessed the IL-6 serum concentration through the ELISA method, the expression of IL-6 in tumor and in immune cells by immunohistochemical and double immunofluorescence staining, the MSI status by immunоhistochemistry for 4 mismatch repair (MMR) proteins, and the genotype distributions for IL6 -174G/C (rs1800795) single-nucleotide polymorphism through PCR-RFLP method. Our results showed that serum IL-6 level were increased in CRC patients as compared with healthy controls (P<0.0001), and in patients with cancers with advanced histologic type (type IV). However, the higher concentration (above the median of 55.71 pg/mL) was with borderline association with longer survival of the patients after surgical therapy (P=0.055, Log rank test). We also found that IL-6+ immune cells prevailed in the invasive front (IF) of tumors compared with the tumor stroma (TS) (P<0.0001). More IL-6+ cells were recruited in the tumors with less advanced histologic type (I+II), with stronger inflammatory infiltrate in the IF, in early pTNM stages (I+II), without lymph node and distant metastases and the higher levels of IL-6+ cells, especially in the IF, were associated with longer survival (P=0.012). The results of our study suggest that although the serum levels of IL-6 are higher in CRC, the increased IL-6+ cells in tumor microenvironment, both in the invasive front and in tumor stroma, as well as the higher serum levels are associated with good prognostic variables and longer survival of the patients mainly in the early stages of CRC.
Collapse
Affiliation(s)
- Maya Gulubova
- Department of General and Clinical Pathology, Forensic Medicine and Deontology
- Department of Surgery
| | - Dimitur Chonov
- Department of Medical Chemistry and Biochemistry, Medical Faculty, Trakia University, Stara Zagora, Bulgaria
| | - Elina Aleksandrova
- Department of Anatomy, Histology and Embriology, Pathology, Latin language, Forensic Medicine and Deontology, Faculty of Medicine, University Prof. Dr. Assen Zlatarov, Burgas
| | - Koni Ivanova
- Department of General and Clinical Pathology, Forensic Medicine and Deontology
| | | | - Tatyana Vlaykova
- Department of Anatomy, Histology and Embriology, Pathology, Latin language, Forensic Medicine and Deontology, Faculty of Medicine, University Prof. Dr. Assen Zlatarov, Burgas
- Department of Medical Biochemistry, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
9
|
Chen X, Ding Y, Yi Y, Chen Z, Fu J, Chang Y. Review of Animal Models of Colorectal Cancer in Different Carcinogenesis Pathways. Dig Dis Sci 2024; 69:1583-1592. [PMID: 38526618 DOI: 10.1007/s10620-024-08384-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 03/05/2024] [Indexed: 03/27/2024]
Abstract
Colorectal cancer (CRC) is a common malignant tumor of the gastrointestinal tract with increasing morbidity and mortality. Exploring the factors affecting colorectal carcinogenesis and controlling its occurrence at its root is as important as studying post-cancer treatment and management. Establishing ideal animal models of CRC is crucial, which can occur through various pathways, such as adenoma-carcinoma sequence, inflammation-induced carcinogenesis, serrated polyp pathway and de-novo pathway. This article aims to categorize the existing well-established CRC animal models based on different carcinogenesis pathways, and to describe their mechanisms, methods, advantages and limitations using domestic and international literature sources. This will provide suggestions for the selection of animal models in early-stage CRC research.
Collapse
Affiliation(s)
- Xue Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Yirong Ding
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Yun Yi
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Zhishan Chen
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Jiaping Fu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
- Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China.
| |
Collapse
|
10
|
Wei X, Dai J, Liu R, Wan G, Gu S, Du Y, Yang X, Wang L, Huang Y, Chen P, Chen X, Yang X, Wang Q. S/O/W Emulsion with CAPE Ameliorates DSS-Induced Colitis by Regulating NF-κB Pathway, Gut Microbiota and Fecal Metabolome in C57BL/6 Mice. Nutrients 2024; 16:1145. [PMID: 38674835 PMCID: PMC11054280 DOI: 10.3390/nu16081145] [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: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammatory bowel disease (IBD) has attracted much attention worldwide due to its prevalence. In this study, the effect of a solid-in-oil-in-water (S/O/W) emulsion with Caffeic acid phenethyl ester (CAPE, a polyphenolic active ingredient in propolis) on dextran sulfate sodium (DSS)-induced colitis in C57BL/6 mice was evaluated. The results showed that CAPE-emulsion could significantly alleviate DSS-induced colitis through its effects on colon length, reduction in the disease activity index (DAI), and colon histopathology. The results of ELISA and Western blot analysis showed that CAPE-emulsion can down-regulate the excessive inflammatory cytokines in colon tissue and inhibit the expression of p65 in the NF-κB pathway. Furthermore, CAPE-emulsion promoted short-chain fatty acids production in DSS-induced colitis mice. High-throughput sequencing results revealed that CAPE-emulsion regulates the imbalance of gut microbiota by enhancing diversity, restoring the abundance of beneficial bacteria (such as Odoribacter), and suppressing the abundance of harmful bacteria (such as Afipia, Sphingomonas). The results of fecal metabolome showed that CAPE-emulsion restored the DSS-induced metabolic disorder by affecting metabolic pathways related to inflammation and cholesterol metabolism. These research results provide a scientific basis for the use of CPAE-emulsions for the development of functional foods for treating IBD.
Collapse
Affiliation(s)
- Xuelin Wei
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Juan Dai
- School of Laboratory Medicine, Chengdu Medical College, Chengdu 610500, China;
| | - Ruijia Liu
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Guochao Wan
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Shiyu Gu
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Yuwei Du
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Xinyue Yang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Lijun Wang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Yukun Huang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Pengfei Chen
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
| | - Xianggui Chen
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China
| | - Xiao Yang
- School of Food and Bioengineering, Xihua University, Chengdu 610039, China; (X.W.); (R.L.); (G.W.); (S.G.); (Y.D.); (X.Y.); (L.W.); (Y.H.); (P.C.); (X.C.)
- Chongqing Key Laboratory of Specialty Food Co-Built by Sichuan and Chongqing, Chengdu 610039, China
| | - Qin Wang
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
11
|
Wang Y, Gao JZ, Sakaguchi T, Maretzky T, Gurung P, Narayanan NS, Short S, Xiong Y, Kang Z. LRRK2 G2019S Promotes Colon Cancer Potentially via LRRK2-GSDMD Axis-Mediated Gut Inflammation. Cells 2024; 13:565. [PMID: 38607004 PMCID: PMC11011703 DOI: 10.3390/cells13070565] [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: 02/20/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/13/2024] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) is a serine-threonine protein kinase belonging to the ROCO protein family. Within the kinase domain of LRRK2, a point mutation known as LRRK2 G2019S has emerged as the most prevalent variant associated with Parkinson's disease. Recent clinical studies have indicated that G2019S carriers have an elevated risk of cancers, including colon cancer. Despite this observation, the underlying mechanisms linking LRRK2 G2019S to colon cancer remain elusive. In this study, employing a colitis-associated cancer (CAC) model and LRRK2 G2019S knock-in (KI) mouse model, we demonstrate that LRRK2 G2019S promotes the pathogenesis of colon cancer, characterized by increased tumor number and size in KI mice. Furthermore, LRRK2 G2019S enhances intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, KI mice exhibit heightened susceptibility to DSS-induced colitis, with inhibition of LRRK2 kinase activity ameliorating colitis severity and CAC progression. Our investigation also reveals that LRRK2 G2019S promotes inflammasome activation and exacerbates gut epithelium necrosis in the colitis model. Notably, GSDMD inhibitors attenuate colitis in LRRK2 G2019S KI mice. Taken together, our findings offer experimental evidence indicating that the gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, suggesting LRRK2 as a potential therapeutic target in colon cancer patients exhibiting hyper LRRK2 kinase activity.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Joyce Z. Gao
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Taylor Sakaguchi
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Thorsten Maretzky
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Prajwal Gurung
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Nandakumar S. Narayanan
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA 52242, USA
- Department of Neurology, University of Iowa, Iowa City, IA 52242, USA
| | - Sarah Short
- Department of Internal Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Yiqin Xiong
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Zizhen Kang
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
12
|
Cossu AM, Melisi F, Noviello TMR, Pasquale LS, Grisolia P, Reale C, Bocchetti M, Falco M, Tammaro C, Accardo N, Longo F, Allosso S, Mesolella M, Addeo R, Perri F, Ottaiano A, Ricciardiello F, Amler E, Ambrosino C, Misso G, Ceccarelli M, Caraglia M, Scrima M. MiR-449a antagonizes EMT through IL-6-mediated trans-signaling in laryngeal squamous cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102140. [PMID: 38425711 PMCID: PMC10901858 DOI: 10.1016/j.omtn.2024.102140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024]
Abstract
MicroRNAs (miRNAs) are involved in post-transcriptional gene expression regulation and in mechanisms of cancer growth and metastases. In this light, miRNAs could be promising therapeutic targets and biomarkers in clinical practice. Therefore, we investigated if specific miRNAs and their target genes contribute to laryngeal squamous cell carcinoma (LSCC) development. We found a significant decrease of miR-449a in LSCC patients with nodal metastases (63.3%) compared with patients without nodal involvement (44%). The AmpliSeq Transcriptome of HNO-210 miR-449a-transfected cell lines allowed the identification of IL6-R as a potential target. Moreover, the downregulation of IL6-R and the phosphorylation reduction of the downstream signaling effectors, suggested the inhibition of the IL-6 trans-signaling pathway. These biochemical effects were paralleled by a significant inhibition of invasion and migration in vitro and in vivo, supporting an involvement of epithelial-mesenchymal transition. These findings indicate that miR-449a contributes to suppress the metastasization of LSCC by the IL-6 trans-signaling block and affects sensitivity to external stimuli that mimic pro-inflammatory conditions.
Collapse
Affiliation(s)
- Alessia Maria Cossu
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Federica Melisi
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Teresa Maria Rosaria Noviello
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Napoli, Italy
| | - Lucia Stefania Pasquale
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Piera Grisolia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Carla Reale
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Marco Bocchetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Michela Falco
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Chiara Tammaro
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Nunzio Accardo
- Ear, Nose, and Throat Unit, AORN "Antonio Cardarelli", Naples, Italy
| | - Francesco Longo
- Head and Neck Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Salvatore Allosso
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, UOC Federico II, 80121 Naples, Italy
| | - Massimo Mesolella
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, UOC Federico II, 80121 Naples, Italy
| | - Raffaele Addeo
- Medical Oncology Unit, San Giovanni di Dio Hospital, 80027 Frattamaggiore, Italy
| | - Francesco Perri
- Head and Neck Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | - Alessandro Ottaiano
- SSD Innovative Therapies for Abdominal Metastases, Abdominal Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, "Fondazione G. Pascale", IRCCS, Naples, Italy
| | | | - Evzen Amler
- UCEEB, Czech Technical University, Třinecká 1024, 273 43 Buštěhrad, Czech
| | - Concetta Ambrosino
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Gabriella Misso
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Michele Ceccarelli
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
- Department of Electrical Engineering and Information Technology, University of Naples "Federico II", Napoli, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| | - Marianna Scrima
- Biogem Scarl, Institute of Genetic Research, 83031 Ariano Irpino, Italy
| |
Collapse
|
13
|
Park EJ, Lee CW. Soluble receptors in cancer: mechanisms, clinical significance, and therapeutic strategies. Exp Mol Med 2024; 56:100-109. [PMID: 38182653 PMCID: PMC10834419 DOI: 10.1038/s12276-023-01150-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 01/07/2024] Open
Abstract
Soluble receptors are soluble forms of receptors found in the extracellular space. They have emerged as pivotal regulators of cellular signaling and disease pathogenesis. This review emphasizes their significance in cancer as diagnostic/prognostic markers and potential therapeutic targets. We provide an overview of the mechanisms by which soluble receptors are generated along with their functions. By exploring their involvement in cancer progression, metastasis, and immune evasion, we highlight the importance of soluble receptors, particularly soluble cytokine receptors and immune checkpoints, in the tumor microenvironment. Although current research has illustrated the emerging clinical relevance of soluble receptors, their therapeutic applications remain underexplored. As the landscape of cancer treatment evolves, understanding and targeting soluble receptors might pave the way for novel strategies for cancer diagnosis, prognosis, and therapy.
Collapse
Affiliation(s)
- Eun-Ji Park
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea.
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon, Republic of Korea.
| |
Collapse
|
14
|
Nagaoka Y, Oshiro K, Yoshino Y, Matsunaga T, Endo S, Ikari A. Activation of the TGF-β1/EMT signaling pathway by claudin-1 overexpression reduces doxorubicin sensitivity in small cell lung cancer SBC-3 cells. Arch Biochem Biophys 2024; 751:109824. [PMID: 37984759 DOI: 10.1016/j.abb.2023.109824] [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: 09/23/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/22/2023]
Abstract
Small-cell lung cancer (SCLC), which accounts for about 15 % of all lung cancers, progresses more rapidly than other histologic types and is rarely detected at an operable early stage. Therefore, chemotherapy, radiation therapy, or their combination are the primary treatments for this type of lung cancer. However, the tendency to acquire resistance to anticancer drugs is a severe problem. Recently, we found that an intercellular adhesion molecule, claudin (CLDN) 1, known to be involved in the migration and invasion of lung cancer cells, is involved in the acquisition of anticancer drug resistance. In the present study, we investigated the effect of CLDN1 on the anticancer-drug sensitivity of SCLC SBC-3 cells. Since epithelial-mesenchymal transition (EMT), which is involved in cancer cell migration and invasion, is well known for its involvement in anticancer-drug sensitivity via inhibition of apoptosis, we also examined EMT involvement in decreased anticancer-drug sensitivity by CLDN1. Sensitivity to doxorubicin (DOX) in SBC-3 cells was significantly decreased by CLDN1 overexpression. CLDN1 overexpression resulted in increased TGF-β1 levels, enhanced EMT induction, and increased migratory potency of SBC-3 cells. The decreased sensitivity of SBC-3 cells to anticancer drugs upon TGF-β1 treatment suggested that activation of the TGF-β1/EMT signaling pathway by CLDN1 causes the decreased sensitivity to anticancer drugs and increased migratory potency. Furthermore, treatments with antiallergic agents tranilast and zoledronic acid, known EMT inhibitors, significantly mitigated the decreased sensitivity of CLDN1-overexpressing SBC-3 cells to DOX. These results suggest that EMT inhibitors might effectively overcome reduced sensitivity to anticancer drugs in CLDN1-overexpressing SCLC cells.
Collapse
Affiliation(s)
- Yuri Nagaoka
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Kotone Oshiro
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Yuta Yoshino
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Laboratory of Bioinformatics, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan; Center for One Medicine Innovative Translational Research (COMIT), Gifu Pharmaceutical University, Gifu 501-1196, Japan.
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| |
Collapse
|
15
|
Garbers C, Lokau J. Cytokines of the interleukin-6 family as emerging targets in inflammatory bowel disease. Expert Opin Ther Targets 2024; 28:57-65. [PMID: 38217849 DOI: 10.1080/14728222.2024.2306341] [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: 10/11/2023] [Accepted: 01/12/2024] [Indexed: 01/15/2024]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) is an umbrella term that includes different chronic inflammatory diseases of the gastrointestinal tract, most commonly Crohn's disease and ulcerative colitis. IBD affects more than 6 million people worldwide and constitutes not only a debilitating disease for the patients, but also a significant factor for society due to costs for health care and reduced working capacity. Despite the introduction of biologicals for the treatment of IBD, the identification of novel targets that could lead to novel therapeutics is still needed. AREAS COVERED In this review, we summarize current knowledge about the interleukin-6 family of cytokines as potential therapeutic targets for improving the therapy of patients with IBD. We discuss cytokines like IL-6 itself for which therapeutics such as inhibitory monoclonal antibodies have already entered the clinics, but also focus on other family members whose therapeutic potential has not been explored yet. EXPERT OPINION The different cytokines of the IL-6 family offer multiple therapeutic targets that can potentially be used to treat patients with inflammatory bowel disease, but unwanted side effects like inhibition of epithelial regeneration have to be considered.
Collapse
Affiliation(s)
- Christoph Garbers
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Juliane Lokau
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Liu JY, Tsai FL, Chuang YL, Ye JC. Aqueous extracts of Ocimum gratissimum mitigate colitis and protect against AOM/DSS-induced colorectal cancer in mice. Carcinogenesis 2023; 44:837-846. [PMID: 37864831 DOI: 10.1093/carcin/bgad074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023] Open
Abstract
In this study, we explored the in vivo effects of Ocimum gratissimum aqueous extracts (OGE) on colorectal cancer (CRC) development provoked by azoxymethane/dextran sodium sulfate (AOM/DSS). The results showed a significant reduction in the tumor load and tumor number for the OGEH group that received continued administration of OGE compared to the AOM/DSS group, with P values of <0.01, but this was not observed in the OGEHs group that received separated administration of OGE. All groups except the control group exhibited aberrant crypt foci (ACF) and adenocarcinoma of lesion pathology in colon, and both conditions were significantly reduced in the OGEH group (P < 0.01) as compared to the AOM/DSS group. Subsequent investigation into whether OGE exhibits eliminative effects on DSS-induced severe colitis (SC) in mice showed that the disease activity index score was significantly reduced in the OGE-treated groups (P < 0.01), also colon colitis histological score was reversed. These data suggest that OGE may be potentially effective in preventing CRC when administered throughout the promotional stages of carcinogenesis by inhibiting inflammatory SC.
Collapse
Affiliation(s)
- Jer-Yuh Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Fang-Ling Tsai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Ya-Ling Chuang
- Animal Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Je-Chiuan Ye
- Department of Bachelor's Degree Program for Indigenous Peoples in Senior Health and Care Management, National Taitung University, Taitung, Taiwan
- Master Program in Biomedical Science, National Taitung University, Taitung, Taiwan
| |
Collapse
|
17
|
Napiórkowska M, Kurpios-Piec D, Kiernozek-Kalińska E, Leśniak A, Klawikowska M, Bujalska-Zadrożny M. New aryl-/heteroarylpiperazine derivatives of 1,7-dimethyl-8,9-diphenyl-4-azatricyclo[5.2.1.0 2,6]dec-8-ene-3,5,10-trione: Synthesis and preliminary studies of biological activities. Bioorg Med Chem 2023; 96:117518. [PMID: 37951135 DOI: 10.1016/j.bmc.2023.117518] [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: 08/28/2023] [Revised: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/13/2023]
Abstract
Compounds containing dicarboximide skeleton such as succinimides, maleimides, glutarimides, and phthalimides possess broad biological properties including anti-fungal, antibacterial, antidepressant, or analgesic activities. The piperazine ring is found in a wide range of molecules that have demonstrated a variety of biological functions such as anticancer action and 5-HT receptors agonist/antagonist activity. In the present study, we combined both structures to develop new antitumor agents, a series of piperazine derivatives of 1,7-dimethyl-8,9-diphenyl-4-azatricyclo[5.2.1.02,6]dec-8-ene-3,5,10-trione and evaluated their biological activity. The structures of all tested compounds were confirmed by 1H and 13C NMR and by ESI MS spectral analysis. Their cytotoxicity was assessed in vitro against eight human cancer cell lines, namely prostate (PC3), colon (HCT116, SW480, SW620), leukemia (K562), liver (HepG2), lung (A549) and breast (MDA-Mb-231) in contrast to normal HMEC-1 cell line, by using MTT and Trypan blue method. The tested compounds showed significant activity toward cancer cells. The most pronounced cytotoxic effect was observed in K562 and HCT116 with IC50 values below 10 μM for all studied compounds. Importantly, the most promising derivatives for each cancer cell line (IC50 < 10 μM) exerted a weaker cytotoxic effect toward normal HMEC-1 cells than cancer cells. The evaluation of proapoptotic and inhibitory effects on IL-6 release showed that K562 and HCT116 cells were more sensitive to studied compounds than other cancer cell lines. Furthermore, for all piperazine derivatives, the functional activities at the 5-HT1A, D2 receptors as well as their binding affinities at the 5-HT2A, H1 and M receptors, were determined. The current investigation was able to successfully design compounds with both serotoninergic and anticancer properties. It serves as a good starting point for a multimodal approach for the management of cancer and cancer-related symptoms.
Collapse
Affiliation(s)
- Mariola Napiórkowska
- Chair and Department of Biochemistry, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland.
| | - Dagmara Kurpios-Piec
- Chair and Department of Biochemistry, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Ewelina Kiernozek-Kalińska
- Department of Immunology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str., 02-096 Warsaw, Poland
| | - Anna Leśniak
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Małgorzata Klawikowska
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| | - Magdalena Bujalska-Zadrożny
- Department of Pharmacodynamics, Faculty of Pharmacy, Centre for Preclinical Research and Technology, Medical University of Warsaw, 1 Banacha Str., 02-097 Warsaw, Poland
| |
Collapse
|
18
|
Glass J, Robinson RL, Greenway G, Jones G, Sharma S. Diabetic Müller-Glial-Cell-Specific Il6ra Knockout Mice Exhibit Accelerated Retinal Functional Decline and Thinning of the Inner Nuclear Layer. Invest Ophthalmol Vis Sci 2023; 64:1. [PMID: 38038619 PMCID: PMC10697173 DOI: 10.1167/iovs.64.15.1] [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: 09/11/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
Purpose Interleukin-6 (IL-6) is implicated in the pathology of diabetic retinopathy (DR). IL-6 trans-signaling via soluble IL-6 receptor (IL-6R) is primarily responsible for its pro-inflammatory functions, whereas cis-signaling via membrane-bound IL-6R is anti-inflammatory. Using a Müller-glial-cell-specific Il6ra-/- mouse, we examined how loss of IL-6 cis-signaling in Müller glial cells (MGCs) affected retinal thinning and electroretinography (ERG) response over 9 months of diabetes. Methods Diabetes was induced in wildtype and knockout mice with streptozotocin (40 mg/kg, daily for 5 days). Spectral domain optical coherence tomography (SD-OCT), ERG, and fundoscopy/fluorescein angiography (FA) were assessed at 2, 6, and 9 months of diabetes. MGCs and bipolar neurons were examined in retinal tissue sections by immunofluorescence. Results Diabetic MGC Il6ra-/- mice had significantly thinner retinas than diabetic wildtype mice at 2 (-7.6 µm), 6 (-12.0 µm), and 9 months (-5.0 µm) of diabetes, as well as significant thinning of the inner nuclear layer (INL). Diabetic MGC Il6ra-/- mice also showed a reduction in scotopic B-wave amplitude and B-wave/A-wave ratio earlier than wildtype diabetic mice. In retinal sections, we found a decrease in bipolar neuronal marker PKCα only in diabetic MGC Il6ra-/- mice, which was significantly lower than both controls and diabetic wildtype mice. Glutamine synthetase, a Müller cell marker, was reduced in both wildtype and MGC Il6ra-/- diabetic mice compared to their respective controls. Conclusions IL-6 cis-signaling in MGCs contributes to maintenance of the INL in diabetes, and loss of the IL-6 receptor reduces MGC-mediated neuroprotection of bipolar neurons in the diabetic retina.
Collapse
Affiliation(s)
- Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Rebekah L. Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Garrett Jones
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, Georgia, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, Georgia, United States
- Department of Ophthalmology, Augusta University, Augusta, Georgia, United States
| |
Collapse
|
19
|
Hoffman JM, Robinson R, Greenway G, Glass J, Budkin S, Sharma S. Blockade of interleukin-6 trans-signaling prevents mitochondrial dysfunction and cellular senescence in retinal endothelial cells. Exp Eye Res 2023; 237:109721. [PMID: 37956941 PMCID: PMC10759313 DOI: 10.1016/j.exer.2023.109721] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Interleukin-6 (IL-6) is a multifaceted cytokine implicated in the pathogenesis of diabetic retinopathy (DR). Its activity extends through cis- and trans-signaling (TS) pathways, with cis-signaling limited to specific cell types possessing the membrane-bound IL-6 receptor, while trans-signaling broadly activates various cells without the membrane bound IL-6 receptor, including retinal endothelial cells. In this study, we determined the effects of interleukin-6 trans-signaling on mitochondrial dysfunction and cellular senescence in human retinal endothelial cells (HRECs). HRECs were cultured and treated with IL-6 + soluble IL-6R or Hyper IL-6 to activate trans-signaling, along with sgp130Fc for inhibition. RT-PCR was used to analyze gene expression changes associated with inflammation and senescence. Cellular senescence was assessed using SA β-gal staining. Mitochondrial function was evaluated using Seahorse XFe24 Bioanalyzer. IL-6 trans-signaling induced inflammatory gene expression as indicated by the upregulation of ICAM1, MCP1, and SERPINA3 levels. Additionally, it reduced mitochondrial respiration and oxidative phosphorylation, and these effects were counteracted by sgp130Fc. Moreover, IL-6 trans-signaling led to altered expression of apoptosis-associated genes, including downregulation of FIS1, BCL2, and MCL1, while promoting cellular senescence, a phenomenon mitigated by sgp130Fc. These results not only deepen our understanding of IL-6 in DR but also carry broader implications for age-related diseases and the aging process itself. This study underscores the potential therapeutic value of targeting IL-6 trans-signaling with sgp130Fc as a promising anti-inflammatory approach for DR and potentially other inflammatory conditions. Further in-vivo investigations are warranted to elucidate the function of IL-6 trans-signaling in aging-related pathologies and overall organismal health.
Collapse
Affiliation(s)
- Jessica M Hoffman
- Department of Biological Sciences, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| | - Rebekah Robinson
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Grace Greenway
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Joshua Glass
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Stepan Budkin
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA; Department of Ophthalmology, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
20
|
Chiriac MT, Hracsko Z, Becker C, Neurath MF. STAT2 Controls Colorectal Tumorigenesis and Resistance to Anti-Cancer Drugs. Cancers (Basel) 2023; 15:5423. [PMID: 38001683 PMCID: PMC10670206 DOI: 10.3390/cancers15225423] [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: 10/10/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Colorectal cancer (CRC) is a significant socioeconomic burden in modern society and is accountable for millions of premature deaths each year. The role of signal transducer and activator of transcription 2 (STAT2)-dependent signaling in this context is not yet fully understood, and no therapies targeting this pathway are currently being pursued. We investigated the role of STAT2 in CRC using experimental mouse models coupled with RNA-sequencing (RNA-Seq) data and functional assays with anti-cancer agents in three-dimensional tumoroids. Stat2-/- mice showed greater resistance to the development of CRC in both inflammation-driven and inflammation-independent experimental CRC models. In ex vivo studies, tumoroids derived from Stat2-/- mice with the multiple intestinal neoplasia (Min) mutant allele of the adenomatous polyposis coli (Apc) locus exhibited delayed growth, were overall smaller and more differentiated as compared with tumoroids from ApcMin/+ wildtype (WT) mice. Notably, tumoroids from ApcMin/+ Stat2-/- mice were more susceptible to anti-cancer agents inducing cell death by different mechanisms. Our findings clearly indicated that STAT2 promotes CRC and suggested that interventions targeting STAT2-dependent signals might become an attractive therapeutic option for patients with CRC.
Collapse
Affiliation(s)
- Mircea T. Chiriac
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Zsuzsanna Hracsko
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Gastroenterology, Endocrinology and Pneumology, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, 91054 Erlangen, Germany
- Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, University of Erlangen-Nuremberg, 91054 Erlangen, Germany
| |
Collapse
|
21
|
Gonzalez-Valdivieso J, Vallejo R, Rodriguez-Rojo S, Santos M, Schneider J, Arias FJ, Girotti A. CD44-targeted nanoparticles for co-delivery of docetaxel and an Akt inhibitor against colorectal cancer. BIOMATERIALS ADVANCES 2023; 154:213595. [PMID: 37639856 DOI: 10.1016/j.bioadv.2023.213595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 07/24/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
New strategies to develop drug-loaded nanocarriers with improved therapeutic efficacy are needed for cancer treatment. Herein we report a novel drug-delivery nanosystem comprising encapsulation of the chemotherapeutic drug docetaxel (DTX) and recombinant fusion of a small peptide inhibitor of Akt kinase within an elastin-like recombinamer (ELR) vehicle. This combined approach is also precisely targeted to colorectal cancer cells by means of a chemically conjugated DNA aptamer specific for the CD44 tumor marker. This 53 nm dual-approach nanosystem was found to selectively affect cell viability (2.5 % survival) and proliferation of colorectal cancer cells in vitro compared to endothelial cells (50 % survival), and to trigger both apoptosis- and necrosis-mediated cell death. Our findings also show that the nanohybrid particles remain stable under physiological conditions, trigger sustained drug release and possess an adequate pharmacokinetic profile after systemic intravenous administration. In vivo assays showed that these dual-approach nanohybrids significantly reduced the number of tumor polyps along the colorectal tract in a murine colorectal cancer model. Furthermore, systemic administration of advanced nanohybrids induced tissue recovery by improving the morphology of gastrointestinal crypts and the tissue architecture. Taken together, these findings indicate that our strategy of an advanced dual-approach nanosystem allows us to achieve successful controlled release of chemotherapeutics in cancer cells and may have a promising potential for colorectal cancer treatment.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain
| | - Reinaldo Vallejo
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Soraya Rodriguez-Rojo
- BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE Research Group (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Valladolid, Spain
| | - Jose Schneider
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Department of Obstetrics & Gynecology, University of Valladolid, School of Medicine, Valladolid, Spain
| | - Francisco Javier Arias
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| | - Alessandra Girotti
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| |
Collapse
|
22
|
Monsour M, Croci DM, Grüter BE, Taussky P, Marbacher S, Agazzi S. Cerebral Aneurysm and Interleukin-6: a Key Player in Aneurysm Generation and Rupture or Just One of the Multiple Factors? Transl Stroke Res 2023; 14:631-639. [PMID: 36042111 DOI: 10.1007/s12975-022-01079-4] [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: 05/16/2022] [Revised: 08/08/2022] [Accepted: 08/25/2022] [Indexed: 10/14/2022]
Abstract
Intracranial aneurysm (IA) rupture is a common cause of subarachnoid hemorrhage (SAH) with high mortality and morbidity. Inflammatory interleukins (IL), such as IL-6, play an important role in the occurrence and rupture of IA causing SAH. With this review we aim to elucidate the specific role of IL-6 in aneurysm formation and rupture in preclinical and clinical studies. IL-6 is a novel cytokine in that it has pro-inflammatory and anti-inflammatory signaling pathways. In preclinical and clinical studies of IA formation, elevated and reduced levels of IL-6 are reported. Poor post-rupture prognosis and increased rupture risk, however, are associated with higher levels of IL-6. By better understanding the relationships between IL-6 and IA formation and rupture, IL-6 may serve as a biomarker in high-risk populations. Furthermore, by better understanding the IL-6 signaling mechanisms in IA formation and rupture, IL-6 may optimize surveillance and treatment strategies. This review examines the association between IL-6 and IA, while also suggesting future research directions.
Collapse
Affiliation(s)
- Molly Monsour
- Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - Davide Marco Croci
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Basil E Grüter
- Program for Regenerative Neuroscience, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Neurosurgery, Kantonsspital Aarau, c/o NeuroResearch Office, Tellstrasse 1, 5001, Aarau, Switzerland
| | - Philipp Taussky
- Department of Neurosurgery, Clinical Neurosciences Center, University of Utah, 175 N Medical Drive East, Salt Lake City, UT, 84132, USA
| | - Serge Marbacher
- Program for Regenerative Neuroscience, Department for BioMedical Research, University of Bern, Bern, Switzerland
- Department of Neurosurgery, Kantonsspital Aarau, c/o NeuroResearch Office, Tellstrasse 1, 5001, Aarau, Switzerland
| | - Siviero Agazzi
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
23
|
Schumacher N, Thomsen I, Brundert F, Hejret V, Düsterhöft S, Tichý B, Schmidt-Arras D, Voss M, Rose-John S. EGFR stimulation enables IL-6 trans-signalling via iRhom2-dependent ADAM17 activation in mammary epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119489. [PMID: 37271223 DOI: 10.1016/j.bbamcr.2023.119489] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/14/2023] [Accepted: 05/05/2023] [Indexed: 06/06/2023]
Abstract
The cytokine interleukin-6 (IL-6) has considerable pro-inflammatory properties and is a driver of many physiological and pathophysiological processes. Cellular responses to IL-6 are mediated by membrane-bound or soluble forms of the IL-6 receptor (IL-6R) complexed with the signal-transducing subunit gp130. While expression of the membrane-bound IL-6R is restricted to selected cell types, soluble IL-6R (sIL-6R) enables gp130 engagement on all cells, a process termed IL-6 trans-signalling and considered to be pro-inflammatory. sIL-6R is predominantly generated through proteolytic processing by the metalloproteinase ADAM17. ADAM17 also liberates ligands of the epidermal growth factor receptor (EGFR), which is a prerequisite for EGFR activation and results in stimulation of proliferative signals. Hyperactivation of EGFR mostly due to activating mutations drives cancer development. Here, we reveal an important link between overshooting EGFR signalling and the IL-6 trans-signalling pathway. In epithelial cells, EGFR activity induces not only IL-6 expression but also the proteolytic release of sIL-6R from the cell membrane by increasing ADAM17 surface activity. We find that this derives from the transcriptional upregulation of iRhom2, a crucial regulator of ADAM17 trafficking and activation, upon EGFR engagement, which results in increased surface localization of ADAM17. Also, phosphorylation of the EGFR-downstream mediator ERK mediates ADAM17 activity via interaction with iRhom2. In sum, our study reveals an unforeseen interplay between EGFR activation and IL-6 trans-signalling, which has been shown to be fundamental in inflammation and cancer.
Collapse
Affiliation(s)
- Neele Schumacher
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany.
| | - Ilka Thomsen
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Florian Brundert
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Vaclav Hejret
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | - Stefan Düsterhöft
- Institute of Molecular Pharmacology, University Hospital Aachen/RWTH, Aachen, Germany
| | - Boris Tichý
- CEITEC-Central European Institute of Technology, Masaryk University, Czech Republic
| | | | - Matthias Voss
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Medical Faculty, Kiel University, Germany
| |
Collapse
|
24
|
Rose-John S, Jenkins BJ, Garbers C, Moll JM, Scheller J. Targeting IL-6 trans-signalling: past, present and future prospects. Nat Rev Immunol 2023; 23:666-681. [PMID: 37069261 PMCID: PMC10108826 DOI: 10.1038/s41577-023-00856-y] [Citation(s) in RCA: 101] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2023] [Indexed: 04/19/2023]
Abstract
Interleukin-6 (IL-6) is a key immunomodulatory cytokine that affects the pathogenesis of diverse diseases, including autoimmune diseases, chronic inflammatory conditions and cancer. Classical IL-6 signalling involves the binding of IL-6 to the membrane-bound IL-6 receptor α-subunit (hereafter termed 'mIL-6R') and glycoprotein 130 (gp130) signal-transducing subunit. By contrast, in IL-6 trans-signalling, complexes of IL-6 and the soluble form of IL-6 receptor (sIL-6R) signal via membrane-bound gp130. A third mode of IL-6 signalling - known as cluster signalling - involves preformed complexes of membrane-bound IL-6-mIL-6R on one cell activating gp130 subunits on target cells. Antibodies and small molecules have been developed that block all three forms of IL-6 signalling, but in the past decade, IL-6 trans-signalling has emerged as the predominant pathway by which IL-6 promotes disease pathogenesis. The first selective inhibitor of IL-6 trans-signalling, sgp130, has shown therapeutic potential in various preclinical models of disease and olamkicept, a sgp130Fc variant, had promising results in phase II clinical studies for inflammatory bowel disease. Technological developments have already led to next-generation sgp130 variants with increased affinity and selectivity towards IL-6 trans-signalling, along with indirect strategies to block IL-6 trans-signalling. Here, we summarize our current understanding of the biological outcomes of IL-6-mediated signalling and the potential for targeting this pathway in the clinic.
Collapse
Affiliation(s)
- Stefan Rose-John
- Biochemical Institute, Medical Faculty, Christian-Albrechts-University, Kiel, Germany
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, Australia
| | - Christoph Garbers
- Department of Pathology, Otto-von-Guericke-University Magdeburg, Medical Faculty, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation (GC:I3), Otto-von-Guericke-University, Magdeburg, Germany
- Center for Health and Medical Prevention (CHaMP), Otto-von-Guericke-University, Magdeburg, Germany
| | - Jens M Moll
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany.
| |
Collapse
|
25
|
Chen Y, Zhou J, Xu S, Nie J. Role of Interleukin-6 Family Cytokines in Organ Fibrosis. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:239-253. [PMID: 37900004 PMCID: PMC10601952 DOI: 10.1159/000530288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/02/2023] [Indexed: 10/31/2023]
Abstract
Background Organ fibrosis remains an important cause of high incidence rate and mortality worldwide. The prominent role of interleukin-6 (IL-6) family members represented by IL-6 in inflammation has been extensively studied, and drugs targeting IL-6 have been used clinically. Because of the close relationship between inflammation and fibrosis, researches on the role of IL-6 family members in organ fibrosis are also gradually emerging. Summary In this review, we systematically reviewed the role of IL-6 family members in fibrosis and their possible mechanisms. We listed the role of IL-6 family members in organ fibrosis and drew two diagrams to illustrate the downstream signal transductions of IL-6 family members. We also summarized the effect of some IL-6 family members' antagonists in a table. Key Messages Fibrosis contributes to organ structure damage, organ dysfunction, and eventually organ failure. Although IL-6 family cytokines have similar downstream signal pathways, different members play various roles in an organ-specific manner which might be partly due to their different target cell populations. The pathogenic role of individual member in various diseases needs to be deciphered carefully.
Collapse
Affiliation(s)
- Ying Chen
- Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiaxin Zhou
- Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shihui Xu
- Department of Allergy, Immunology and Rheumatology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jing Nie
- Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Jiang Y, Zhang J, Shi C, Li X, Jiang Y, Mao R. NF- κB: a mediator that promotes or inhibits angiogenesis in human diseases? Expert Rev Mol Med 2023; 25:e25. [PMID: 37503730 DOI: 10.1017/erm.2023.20] [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] [Indexed: 07/29/2023]
Abstract
The nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) signaling pathway, which is conserved in invertebrates, plays a significant role in human diseases such as inflammation-related diseases and carcinogenesis. Angiogenesis refers to the growth of new capillary vessels derived from already existing capillaries and postcapillary venules. Maintaining normal angiogenesis and effective vascular function is a prerequisite for the stability of organ tissue function, and abnormal angiogenesis often leads to a variety of diseases. It has been suggested that NK-κB signalling molecules under pathological conditions play an important role in vascular differentiation, proliferation, apoptosis and tumourigenesis by regulating the transcription of multiple target genes. Many NF-κB inhibitors are being tested in clinical trials for cancer treatment and their effect on angiogenesis is summarised. In this review, we will summarise the role of NF-κB signalling in various neovascular diseases, especially in tumours, and explore whether NF-κB can be used as an attack target or activation medium to inhibit tumour angiogenesis.
Collapse
Affiliation(s)
- Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, 30Tongyang North Road, Pingchao Town, Nantong 226361, Jiangsu, People's Republic of China
| | - Conglin Shi
- Department of Pathogenic Biology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Xingjuan Li
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong 226001, Jiangsu, People's Republic of China
| |
Collapse
|
27
|
Wang Y, Gao JZ, Sakaguchi T, Maretzky T, Gurung P, Short S, Xiong Y, Kang Z. LRRK2 G2019S promotes the development of colon cancer via modulating intestinal inflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546897. [PMID: 37425755 PMCID: PMC10326997 DOI: 10.1101/2023.06.28.546897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
LRRK2 G2019S is the most prevalent variant associated with Parkinson's disease (PD), found in 1-3% of sporadic and 4-8% of familial PD cases. Intriguingly, emerging clinical studies have suggested that LRRK2 G2019S carriers have an increased risk of cancers including colorectal cancer. However, the underlying mechanisms of the positive correlation between LRRK2-G2019S and colorectal cancer remain unknown. Using a mouse model of colitis-associated cancer (CAC) and LRRK2 G2019S knockin (KI) mice, here we report that LRRK2 G2019S promotes the pathogenesis of colon cancer as evidenced by increased tumor number and tumor size in LRRK2 G2019S KI mice. LRRK2 G2019S promoted intestinal epithelial cell proliferation and inflammation within the tumor microenvironment. Mechanistically, we found that LRRK2 G2019S KI mice are more susceptible to dextran sulfate sodium (DSS)-induced colitis. Suppressing the kinase activity of LRRK2 ameliorated the severity of colitis in both LRRK2 G2019S KI and WT mice. At the molecular level, our investigation unveiled that LRRK2 G2019S promotes the production of reactive oxygen species, triggers inflammasome activation, and induces cell necrosis in the gut epithelium in a mouse model of colitis. Collectively, our data provide direct evidence that gain-of-kinase activity in LRRK2 promotes colorectal tumorigenesis, implicating LRRK2 as a potential target in colon cancer patients with hyper LRRK2 kinase activity.
Collapse
|
28
|
Pei L, Liu Y, Liu L, Gao S, Gao X, Feng Y, Sun Z, Zhang Y, Wang C. Roles of cancer-associated fibroblasts (CAFs) in anti- PD-1/PD-L1 immunotherapy for solid cancers. Mol Cancer 2023; 22:29. [PMID: 36759842 PMCID: PMC9912573 DOI: 10.1186/s12943-023-01731-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
In recent years, breakthroughs have been made in tumor immunotherapy. However, tumor immunotherapy, particularly anti-PD-1/PD-L1 immune checkpoint inhibitors, is effective in only a small percentage of patients in solid cancer. How to improve the efficiency of cancer immunotherapy is an urgent problem to be solved. As we all know, the state of the tumor microenvironment (TME) is an essential factor affecting the effectiveness of tumor immunotherapy, and the cancer-associated fibroblasts (CAFs) in TME have attracted much attention in recent years. As one of the main components of TME, CAFs interact with cancer cells and immune cells by secreting cytokines and vesicles, participating in ECM remodeling, and finally affecting the immune response process. With the in-depth study of CAFs heterogeneity, new strategies are provided for finding targets of combination immunotherapy and predicting immune efficacy. In this review, we focus on the role of CAFs in the solid cancer immune microenvironment, and then further elaborate on the potential mechanisms and pathways of CAFs influencing anti-PD-1/PD-L1 immunotherapy. In addition, we summarize the potential clinical application value of CAFs-related targets and markers in solid cancers.
Collapse
Affiliation(s)
- Liping Pei
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China ,grid.412633.10000 0004 1799 0733Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yang Liu
- grid.414008.90000 0004 1799 4638Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, 450008 China
| | - Lin Liu
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Shuochen Gao
- grid.412633.10000 0004 1799 0733Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xueyan Gao
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yudi Feng
- grid.412633.10000 0004 1799 0733Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhenqiang Sun
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yan Zhang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Chengzeng Wang
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
29
|
Sil S, Bertilla J, Rupachandra S. A comprehensive review on RNA interference-mediated targeting of interleukins and its potential therapeutic implications in colon cancer. 3 Biotech 2023; 13:18. [PMID: 36568500 PMCID: PMC9768089 DOI: 10.1007/s13205-022-03421-x] [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: 04/18/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Colon cancer is the world's fourth leading cause of death. It is cancer of the latter part of the large intestine, i.e. the colon. Chronic inflammation over a long period also leads to the development of cancer. Cancer in the colon region is arduous to diagnose and is detected at a later stage when it metastasizes to other parts of the body like the liver, lungs, peritoneum, etc. Colon cancer is a great example of solid tumours associated with chronic inflammation. Although conventional therapies are effective, they lose their effectiveness beyond a certain point. Relapse of the disease occurs frequently. RNA interference (RNAi) is emerging as a great tool to specifically attack the cancer cells of a target site like the colon. RNAi deals with epigenetic changes made in the defective cells which ultimately leads to their death without harming the healthy cells. In this review, two types of epigenetic modulators have been considered, namely siRNA and miRNA, and their effect on interleukins. Interleukins, a class of cytokines, are major inflammatory responses of the body that are released by immune cells like leukocytes and macrophages. Some of these interleukins are pro-inflammatory, thereby promoting inflammation which eventually causes cancer. RNAi can prevent colon cancer by inhibiting pro-inflammatory interleukins.
Collapse
Affiliation(s)
- Sagari Sil
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - Janet Bertilla
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| | - S. Rupachandra
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu, Tamil Nadu 603 203 India
| |
Collapse
|
30
|
Tabikhanova LE, Osipova LP, Churkina TV, Kovalev SS, Filipenko ML, Voronina EN. Increased Frequencies of the ‒174G and ‒572C IL6 Alleles in Populations of Indigenous Peoples of Siberia Compared to Russians. Mol Biol 2023; 57:329-337. [PMID: 37128211 PMCID: PMC10131552 DOI: 10.1134/s002689332302019x] [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: 07/27/2022] [Revised: 09/16/2022] [Accepted: 09/29/2022] [Indexed: 05/03/2023]
Abstract
Abstract-The study of immune response and inflammation gene polymorphisms in a genogeographic context is relevant in the study of human populations. Here, in the indigenous populations of Siberia the frequencies of polymorphic variants ‒174G/C (rs1800795) and ‒572C/G (rs1800796) of the IL6 gene encoding the proinflammatory cytokine IL-6 were determined. For the first time, it was shown that the frequencies of the ‒174G and ‒572C alleles, which determine increased inflammatory response and are also associated with several diseases were statistically significantly higher in ethnic groups of Buryats, Teleuts, Yakuts, Dolgans and Tuvinians than in Russians living in Siberia. These values were in the intermediate position between those in the European and East-Asian groups. We hypothesize an adaptive role of these IL6 genetic variants in human settlement from Africa to the Eurasian continent. However, due to the departure from the traditional way of life and the increasing anthropogenic environmental pollution, the risk of diseases whose pathogenesis is based on inflammation in indigenous Siberian populations is likely increased.
Collapse
Affiliation(s)
- L. E. Tabikhanova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - L. P. Osipova
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - T. V. Churkina
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - S. S. Kovalev
- Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - M. L. Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - E. N. Voronina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
31
|
Gerlach K, Popp V, Wirtz S, Al-Saifi R, Gonzalez Acera M, Atreya R, Dregelies T, Vieth M, Fichtner-Feigl S, McKenzie ANJ, Rosenbauer F, Weigmann B, Neurath MF. PU.1-driven Th9 Cells Promote Colorectal Cancer in Experimental Colitis Models Through Il-6 Effects in Intestinal Epithelial Cells. J Crohns Colitis 2022; 16:1893-1910. [PMID: 35793807 DOI: 10.1093/ecco-jcc/jjac097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Colorectal cancer [CRC] is one of the most frequent malignancies, but the molecular mechanisms driving cancer growth are incompletely understood. We characterised the roles of the cytokine IL-9 and Th9 cells in regulating CRC development. METHODS CRC patient samples and samples from AOM/DSS treated mice were analysed for expression of IL-9, CD3, and PU.1 by FACS analysis and immunohistochemistry. IL-9 citrine reporter mice, IL-9 knockout mice, and PU.1 and GATA3 CD4-Cre conditional knockout mice were studied in the AOM/DSS model. DNA minicircles or hyper-IL-6 were used for overexpression of cytokines in vivo. Effects of IL-6 and IL-9 were determined in organoid and T cell cultures. Claudin2/3 expression was studied by western blotting and bacterial translocation by FISH. RESULTS We uncovered a significant expansion of IL-9- and PU.1-expressing mucosal Th9 cells in CRC patients, with particularly high levels in patients with colitis-associated neoplasias. PU.1+ Th9 cells accumulated in experimental colorectal neoplasias. Deficiency of IL-9 or inactivation of PU.1 in T cells led to impaired tumour growth in vivo, suggesting a protumoral role of Th9 cells. In contrast, GATA3 inactivation did not affect Th9-mediated tumour growth. Mechanistically, IL-9 controls claudin2/3 expression and T cell-derived IL-6 production in colorectal tumours. IL-6 abrogated the anti-proliferative effects of IL-9 in epithelial organoids in vivo. IL-9-producing Th9 cells expand in CRC and control IL-6 production by T cells. CONCLUSIONS IL-9 is a crucial regulator of tumour growth in colitis-associated neoplasias and emerges as potential target for therapy.
Collapse
Affiliation(s)
- Katharina Gerlach
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Vanessa Popp
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Ragheed Al-Saifi
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Miguel Gonzalez Acera
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie [DZI], Erlangen, University of Erlangen-Nuremberg, Germany
| | - Theresa Dregelies
- Institute of Pathology, Klinikum Bayreuth, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Stefan Fichtner-Feigl
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Andrew N J McKenzie
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Frank Rosenbauer
- Laboratory of Molecular Stem Cell Biology, University of Münster, Münster, Germany
| | - Benno Weigmann
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany.,Medical Immunology Campus Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Kussmaul Campus for Medical Research, University of Erlangen-Nuremberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie [DZI], Erlangen, University of Erlangen-Nuremberg, Germany
| |
Collapse
|
32
|
Rodriguez Calleja L, Lavaud M, Tesfaye R, Brounais-Le-Royer B, Baud’huin M, Georges S, Lamoureux F, Verrecchia F, Ory B. The p53 Family Members p63 and p73 Roles in the Metastatic Dissemination: Interactions with microRNAs and TGFβ Pathway. Cancers (Basel) 2022; 14:cancers14235948. [PMID: 36497429 PMCID: PMC9741383 DOI: 10.3390/cancers14235948] [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: 10/30/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
TP53 (TP53), p73 (TP73), and p63 (TP63) are members of the p53 transcription factor family, which has many activities spanning from embryonic development through to tumor suppression. The utilization of two promoters and alternative mRNA splicing has been shown to yield numerous isoforms in p53, p63, and p73. TAp73 is thought to mediate apoptosis as a result of nuclear accumulation following chemotherapy-induced DNA damage, according to a number of studies. Overexpression of the nuclear ΔNp63 and ΔNp73 isoforms, on the other hand, suppresses TAp73's pro-apoptotic activity in human malignancies, potentially leading to metastatic spread or inhibition. Another well-known pathway that has been associated to metastatic spread is the TGF pathway. TGFs are a family of structurally related polypeptide growth factors that regulate a variety of cellular functions including cell proliferation, lineage determination, differentiation, motility, adhesion, and cell death, making them significant players in development, homeostasis, and wound repair. Various studies have already identified several interactions between the p53 protein family and the TGFb pathway in the context of tumor growth and metastatic spread, beginning to shed light on this enigmatic intricacy.
Collapse
|
33
|
Marangio A, Biccari A, D’Angelo E, Sensi F, Spolverato G, Pucciarelli S, Agostini M. The Study of the Extracellular Matrix in Chronic Inflammation: A Way to Prevent Cancer Initiation? Cancers (Basel) 2022; 14:cancers14235903. [PMID: 36497384 PMCID: PMC9741172 DOI: 10.3390/cancers14235903] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/04/2022] Open
Abstract
Bidirectional communication between cells and their microenvironment has a key function in normal tissue homeostasis, and in disease initiation, progression and a patient's prognosis, at the very least. The extracellular matrix (ECM), as an element of all tissues and cellular microenvironment, is a frequently overlooked component implicated in the pathogenesis and progression of several diseases. In the inflammatory microenvironment (IME), different alterations resulting from remodeling processes can affect ECM, progressively inducing cancer initiation and the passage toward a tumor microenvironment (TME). Indeed, it has been demonstrated that altered ECM components interact with a variety of surface receptors triggering intracellular signaling that affect cellular pathways in turn. This review aims to support the notion that the ECM and its alterations actively participate in the promotion of chronic inflammation and cancer initiation. In conclusion, some data obtained in cancer research with the employment of decellularized ECM (dECM) models are described. The reported results encourage the application of dECM models to investigate the short circuits contributing to the creation of distinct IME, thus representing a potential tool to avoid the progression toward a malignant lesion.
Collapse
Affiliation(s)
- Asia Marangio
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Andrea Biccari
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Edoardo D’Angelo
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
| | - Francesca Sensi
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Department of Women’s and Children’s Health, University of Padova, 35128 Padova, Italy
| | - Gaya Spolverato
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Marco Agostini
- General Surgery 3, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica, Città della Speranza, 35129 Padova, Italy
- Correspondence: ; Tel.: +39-049-964-0160
| |
Collapse
|
34
|
Cao X, Zhen M, Li L, Wu Z, Zhou C, Huo J, Su S, Xu Y, Jia W, Liao X, Sun Z, Li H, Wang C. Oral fullerene tablets for colorectal cancer therapy based on modulation of tumor inflammatory microenvironments. J Mater Chem B 2022; 10:9457-9465. [PMID: 36346268 DOI: 10.1039/d2tb01518h] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The development and progression of colorectal cancer (CRC) are highly dependent on the long-term inflammatory microenvironment with immune dysregulation in the colorectum. However, effective therapeutics are limited to targeting CRC. Here, we developed oral fullerene tablets (OFTs) that can act directly on the colorectal site by oral administration and reduce the inflammatory state at the tumor site for effective CRC therapy. In detail, OFTs scavenged reactive oxygen species (ROS), restrained the mutation of the wild-type P53, inhibited the activation of the inflammatory pathway nuclear factor-κB (NF-κB) and the signal transducer and activator of transcription 3 (STAT3) in the colorectum of CRC mice. Subsequently, OFTs could greatly reduce the infiltration of pro-inflammatory M1 macrophages and neutrophils at the tumor site, restoring the inflammatory microenvironment and immune homeostasis in the colorectal region, and ultimately achieving the inhibition of CRC. In addition, there were no significant toxic side effects of the long-term administration of OFTs. Our work provides an effective oral therapeutic strategy for CRC therapy by modulating the colorectal tumor inflammatory microenvironment and sheds light on the route for oral nano-materials in the clinical treatment of CRC.
Collapse
Affiliation(s)
- Xinran Cao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingming Zhen
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhanfeng Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Zhou
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiawei Huo
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shenge Su
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan Xu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wang Jia
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaodan Liao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zihao Sun
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Li
- Beijing Fullcan Biotechnology Co. Ltd, Beijing, 100085, China
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China. .,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
35
|
Gamez-Belmonte R, Mahapatro M, Erkert L, Gonzalez-Acera M, Naschberger E, Yu Y, Tena-Garitaonaindia M, Patankar JV, Wagner Y, Podstawa E, Schödel L, Bubeck M, Neurath MF, Stürzl M, Becker C. Epithelial presenilin-1 drives colorectal tumour growth by controlling EGFR-COX2 signalling. Gut 2022; 72:1155-1166. [PMID: 36261293 DOI: 10.1136/gutjnl-2022-327323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/02/2022] [Indexed: 12/08/2022]
Abstract
OBJECTIVE Psen1 was previously characterised as a crucial factor in the pathogenesis of neurodegeneration in patients with Alzheimer's disease. Little, if any, is known about its function in the gut. Here, we uncovered an unexpected functional role of Psen1 in gut epithelial cells during intestinal tumourigenesis. DESIGN Human colorectal cancer (CRC) and control samples were investigated for PSEN1 and proteins of theγ-secretase complex. Tumour formation was analysed in the AOM-DSS and Apc min/+ mouse models using newly generated epithelial-specific Psen1 deficient mice. Psen1 deficient human CRC cells were studied in a xenograft tumour model. Tumour-derived organoids were analysed for growth and RNA-Seq was performed to identify Psen1-regulated pathways. Tumouroids were generated to study EGFR activation and evaluation of the influence of prostanoids. RESULTS PSEN1 is expressed in the intestinal epithelium and its level is increased in human CRC. Psen1-deficient mice developed only small tumours and human cancer cell lines deficient in Psen1 had a reduced tumourigenicity. Tumouroids derived from Psen1-deficient Apc min/+ mice exhibited stunted growth and reduced cell proliferation. On a molecular level, PSEN1 potentiated tumour cell proliferation via enhanced EGFR signalling and COX-2 production. Exogenous administration of PGE2 reversed the slow growth of PSEN1 deficient tumour cells via PGE2 receptor 4 (EP4) receptor signalling. CONCLUSIONS Psen1 drives tumour development by increasing EGFR signalling via NOTCH1 processing, and by activating the COX-2-PGE2 pathway. PSEN1 inhibition could be a useful strategy in treatment of CRC.
Collapse
Affiliation(s)
- Reyes Gamez-Belmonte
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mousumi Mahapatro
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lena Erkert
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Miguel Gonzalez-Acera
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yuqiang Yu
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Duke University Medical Center, Durham, North Carolina, USA
| | | | - Jay V Patankar
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yara Wagner
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Eva Podstawa
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lena Schödel
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marvin Bubeck
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Becker
- Department of Medicine 1, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany .,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
36
|
Qiu Y, Ke S, Chen J, Qin Z, Zhang W, Yuan Y, Meng D, Zhao G, Wu K, Li B, Li D. FOXP3+ regulatory T cells and the immune escape in solid tumours. Front Immunol 2022; 13:982986. [PMID: 36569832 PMCID: PMC9774953 DOI: 10.3389/fimmu.2022.982986] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/01/2022] [Indexed: 01/15/2023] Open
Abstract
FOXP3+ regulatory T (Treg) cells play critical roles in establishing the immunosuppressive tumour microenvironment, which is achieved and dynamically maintained with the contribution of various stromal and immune cell subsets. However, the dynamics of non-lymphoid FOXP3+ Treg cells and the mutual regulation of Treg cells and other cell types in solid tumour microenvironment remains largely unclear. In this review, we summarize the latest findings on the dynamic connections and reciprocal regulations of non-lymphoid Treg cell subsets in accordance with well-established and new emerging hallmarks of cancer, especially on the immune escape of tumour cells in solid tumours. Our comprehension of the interplay between FOXP3+ Treg cells and key hallmarks of cancer may provide new insights into the development of next-generation engineered T cell-based immune treatments for solid tumours.
Collapse
Affiliation(s)
- Yiran Qiu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shouyu Ke
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieqiong Chen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhizhen Qin
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenle Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yaqin Yuan
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dehua Meng
- Department of Orthopedics, Zhongshan Hospital, Fudan University School of Medicine, Shanghai, China
| | - Gang Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejin Wu
- Department of Breast Surgery, Obstetrics and Gynecology Hospital, Fudan University School of Medicine, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Arthritis Research, Guanghua Integrative Medicine Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Integrated TCM & Western Medicine at Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dan Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Respiratory and Critical Care Medicine of Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
37
|
Yu Y, Yue Z, Xu M, Zhang M, Shen X, Ma Z, Li J, Xie X. Macrophages play a key role in tissue repair and regeneration. PeerJ 2022; 10:e14053. [PMID: 36196399 PMCID: PMC9527023 DOI: 10.7717/peerj.14053] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 08/24/2022] [Indexed: 01/19/2023] Open
Abstract
Tissue regeneration after body injury has always been a complex problem to resolve for mammals. In adult mammals, the repair process after tissue injury is often accompanied by continuous and extensive fibrosis, which leads to scars. This process has been shown to severely hinder regeneration. Macrophages, as widely distributed innate immune cells, not only play an important role in various pathological processes, but also participate in the repair process before tissue regeneration and coordinate the regeneration process after repair. This review will discuss the various forms and indispensability of macrophages involved in repair and regeneration, and how macrophages play a role in the repair and regeneration of different tissues.
Collapse
Affiliation(s)
- Yajie Yu
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Zhongyu Yue
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Mengli Xu
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Meiling Zhang
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Xue Shen
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Zihan Ma
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Juan Li
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| | - Xin Xie
- College of Life Science, Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
38
|
Delfini M, Stakenborg N, Viola MF, Boeckxstaens G. Macrophages in the gut: Masters in multitasking. Immunity 2022; 55:1530-1548. [PMID: 36103851 DOI: 10.1016/j.immuni.2022.08.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/17/2022] [Accepted: 08/09/2022] [Indexed: 11/05/2022]
Abstract
The gastrointestinal tract has the important task of absorbing nutrients, a complex process that requires an intact barrier allowing the passage of nutrients but that simultaneously protects the host against invading microorganisms. To maintain and regulate intestinal homeostasis, the gut is equipped with one of the largest populations of macrophages in the body. Here, we will discuss our current understanding of intestinal macrophage heterogeneity and describe their main functions in the different anatomical niches of the gut during steady state. In addition, their role in inflammatory conditions such as infection, inflammatory bowel disease, and postoperative ileus are discussed, highlighting the roles of macrophages in immune defense. To conclude, we describe the interaction between macrophages and the enteric nervous system during development and adulthood and highlight their contribution to neurodegeneration in the context of aging and diabetes.
Collapse
Affiliation(s)
- Marcello Delfini
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Nathalie Stakenborg
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Maria Francesca Viola
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium
| | - Guy Boeckxstaens
- Translational Research Center for GI Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing, KU Leuven-University of Leuven, Leuven, Belgium.
| |
Collapse
|
39
|
Sun X, Ma L, Li X, Wang J, Li Y, Huang Z. Ferulic acid alleviates retinal neovascularization by modulating microglia/macrophage polarization through the ROS/NF-κB axis. Front Immunol 2022; 13:976729. [PMID: 36119027 PMCID: PMC9478033 DOI: 10.3389/fimmu.2022.976729] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation plays a pivotal role in ischemia-induced retinal neovascularization. Targeting microglia/macrophage-based neuroinflammation presents a promising therapeutic strategy. Ferulic acid (FA), a natural and active ingredient in plants, exerts favorable anti-oxidative and anti-inflammatory activities. In this study, we investigated the inhibitory effect of FA against hypoxia-induced retinal angiogenesis using cultured retinal vascular endothelial cells and an oxygen-induced retinopathy mouse (OIR) model. The immunoregulatory effect of FA on microglia/macrophage polarization was evaluated by detecting the expression of specific markers for both pro-inflammatory “M1” and anti-inflammatory “M2” phenotypes using co-immunostaining and polymerase chain reaction assays. The underlying molecular mechanism upon FA treatment was also explored. The results showed that FA supplement markedly inhibited retinal pathological angiogenesis both in vivo and in vitro. In addition, FA switched microglia/macrophage polarization from “M1” towards “M2” phenotype and alleviated the inflammatory response. Mechanically, the anti-angiogenic and anti-inflammatory properties of FA were mainly due to blockade of the ROS/NF-κB pathway. Our data demonstrated an anti-angiogenic effect of FA through regulating M1-to-M2 microglia/macrophage polarization, suggesting a potential therapeutic strategy for retinal neovascular diseases.
Collapse
Affiliation(s)
- Xiaowei Sun
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Lusheng Ma
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Xiao Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiao Wang
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yuanbin Li
- Department of Ophthalmology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Zijing Huang
- Joint Shantou International Eye Center of Shantou University and The Chinese University of Hong Kong, Shantou, China
| |
Collapse
|
40
|
Chan MKK, Chung JYF, Tang PCT, Chan ASW, Ho JYY, Lin TPT, Chen J, Leung KT, To KF, Lan HY, Tang PMK. TGF-β signaling networks in the tumor microenvironment. Cancer Lett 2022; 550:215925. [DOI: 10.1016/j.canlet.2022.215925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 09/05/2022] [Accepted: 09/17/2022] [Indexed: 11/02/2022]
|
41
|
Hashimoto S, Hashimoto A, Muromoto R, Kitai Y, Oritani K, Matsuda T. Central Roles of STAT3-Mediated Signals in Onset and Development of Cancers: Tumorigenesis and Immunosurveillance. Cells 2022; 11:cells11162618. [PMID: 36010693 PMCID: PMC9406645 DOI: 10.3390/cells11162618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/12/2022] [Accepted: 08/20/2022] [Indexed: 02/07/2023] Open
Abstract
Since the time of Rudolf Virchow in the 19th century, it has been well-known that cancer-associated inflammation contributes to tumor initiation and progression. However, it remains unclear whether a collapse of the balance between the antitumor immune response via the immunological surveillance system and protumor immunity due to cancer-related inflammation is responsible for cancer malignancy. The majority of inflammatory signals affect tumorigenesis by activating signal transducer and activation of transcription 3 (STAT3) and nuclear factor-κB. Persistent STAT3 activation in malignant cancer cells mediates extremely widespread functions, including cell growth, survival, angiogenesis, and invasion and contributes to an increase in inflammation-associated tumorigenesis. In addition, intracellular STAT3 activation in immune cells causes suppressive effects on antitumor immunity and leads to the differentiation and mobilization of immature myeloid-derived cells and tumor-associated macrophages. In many cancer types, STAT3 does not directly rely on its activation by oncogenic mutations but has important oncogenic and malignant transformation-associated functions in both cancer and stromal cells in the tumor microenvironment (TME). We have reported a series of studies aiming towards understanding the molecular mechanisms underlying the proliferation of various types of tumors involving signal-transducing adaptor protein-2 as an adaptor molecule that modulates STAT3 activity, and we recently found that AT-rich interactive domain-containing protein 5a functions as an mRNA stabilizer that orchestrates an immunosuppressive TME in malignant mesenchymal tumors. In this review, we summarize recent advances in our understanding of the functional role of STAT3 in tumor progression and introduce novel molecular mechanisms of cancer development and malignant transformation involving STAT3 activation that we have identified to date. Finally, we discuss potential therapeutic strategies for cancer that target the signaling pathway to augment STAT3 activity.
Collapse
Affiliation(s)
- Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
- Correspondence: (S.H.); (T.M.)
| | - Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Ryuta Muromoto
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Yuichi Kitai
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Kenji Oritani
- Department of Hematology, International University of Health and Welfare, Narita 286-8686, Japan
| | - Tadashi Matsuda
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
- Correspondence: (S.H.); (T.M.)
| |
Collapse
|
42
|
Choi SH, Huang AY, Letterio JJ, Kim BG. Smad4-deficient T cells promote colitis-associated colon cancer via an IFN-γ-dependent suppression of 15-hydroxyprostaglandin dehydrogenase. Front Immunol 2022; 13:932412. [PMID: 36045676 PMCID: PMC9420841 DOI: 10.3389/fimmu.2022.932412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Immune cells and the cytokines they produce are important mediators of the transition from colitis to colon cancer, but the mechanisms mediating this disease progression are poorly understood. Interferon gamma (IFN-γ) is known to contribute to the pathogenesis of colitis through immune modulatory mechanisms, and through direct effects on endothelial and epithelial homeostasis. Here we explore whether IFN-γ influences tumor progression by expanding the effector memory T cells (TEM) population and restricting the expression of tumor suppressors in a preclinical model of spontaneous colitis-associated colorectal cancer (CAC). We show that IFN-γ expression is significantly increased both in the T cells and the colonic mucosal epithelia of mice with a T cell-restricted deletion of the TGF-β intermediate, SMAD4 (Smad4TKO). The increase of IFN-γ expression correlates with the onset of spontaneous CAC in Smad4TKO mice by 6 months of age. This phenotype is greatly ameliorated by the introduction of a germline deletion of IFN-γ in Smad4TKO mice (Smad4TKO/IFN-γKO, DKO). DKO mice had a significantly reduced incidence and progression of CAC, and a decrease in the number of mucosal CD4+ TEM cells, when compared to those of Smad4TKO mice. Similarly, the colon epithelia of DKO mice exhibited a non-oncogenic signature with a decrease in the expression of iNOS and p-STAT1, and a restoration of the tumor suppressor gene, 15-hydroxyprostaglandin dehydrogenase (15-PGDH). In vitro, treatment of human colon cancer cells with IFN-γ decreased the expression of 15-PGDH. Our data suggest that Smad4-deficient T cells promote CAC through mechanisms that include an IFN-γ-dependent suppression of the tumor suppressor 15-PGDH.
Collapse
Affiliation(s)
- Sung Hee Choi
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Alex Y. Huang
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals (UH) Rainbow Babies and Children’s Hospital, Cleveland, OH, United States
| | - John J. Letterio
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals (UH) Rainbow Babies and Children’s Hospital, Cleveland, OH, United States
| | - Byung-Gyu Kim
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- *Correspondence: Byung-Gyu Kim,
| |
Collapse
|
43
|
Xue C, Gao Y, Sun Z, Li X, Zhang M, Yang Y, Han Q, Bai C, Zhao RC. Mesenchymal stem cells derived from adipose tissue accelerate the progression of colon cancer by inducing a MTCAF phenotype via ICAM1/STAT3/AKT axis. Front Oncol 2022; 12:837781. [PMID: 36016615 PMCID: PMC9398219 DOI: 10.3389/fonc.2022.837781] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Previous studies have shown that the risk of colon cancer is greatly increased in people with obesity, and fat content in colorectal cancer tissue is increased in people with obesity. As an important part of tumor microenvironment, adipose-derived mesenchymal stem cells (MSCs) are also another important source of cancer-associated fibroblasts (CAFs), which may be one of the important mechanisms of affecting tumor progression. However, the mechanism is poorly defined. In the present study, CAFs were transformed from MSCs [MSC-transformed CAFs (MTCAFs)] by co-culturing with HCT116 cells. Bioinformatics and Western blotting analysis indicated a positive correlation between intercellular adhesion molecule-1(ICAM-1) and the progression of colon cancer. In clinical colon cancer specimens, we found that ICAM-1 was highly expressed and related to shorter disease-free survival, which might act as an indication for the progression of clinical colon cancer. Our data showed that ICAM-1 secreted from MTCAFs could positively promote the proliferation, migration, and invasion of colon cancer cells by activating signal transducer and activator of transcription 3 (STAT3) and Serine/threonine-protein kinase (AKT) signaling and that blocking ICAM-1 in MTCAFs reversed these effects. We further verified that ICAM-1 secreted from MTCAFs promoted tumor progression in vivo. Taken together, ICAM-1 plays a critical role in regulating tumor growth and metastasis, which could be a potential therapeutic target in colon cancer.
Collapse
Affiliation(s)
- Chunling Xue
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Yang Gao
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Zhao Sun
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xuechun Li
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Mingjia Zhang
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
| | - Ying Yang
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Qin Han
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Qin Han, ; Chunmei Bai, ; Robert Chunhua Zhao,
| | - Chunmei Bai
- Department of oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- *Correspondence: Qin Han, ; Chunmei Bai, ; Robert Chunhua Zhao,
| | - Robert Chunhua Zhao
- Beijing Key Laboratory, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Center of Excellence in Tissue Engineering Chinese Academy of Medical Sciences, Peking Union Medical College Hospital, Beijing, China
- *Correspondence: Qin Han, ; Chunmei Bai, ; Robert Chunhua Zhao,
| |
Collapse
|
44
|
Qin W, Luo H, Yang L, Hu D, Jiang SP, Peng DY, Hu JM, Liu SJ. Rubia cordifolia L. ameliorates DSS-induced ulcerative colitis in mice through dual inhibition of NLRP3 inflammasome and IL-6/JAK2/STAT3 pathways. Heliyon 2022; 8:e10314. [PMID: 36082330 PMCID: PMC9445285 DOI: 10.1016/j.heliyon.2022.e10314] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/01/2022] [Accepted: 08/11/2022] [Indexed: 10/25/2022] Open
|
45
|
Shive C, Pandiyan P. Inflammation, Immune Senescence, and Dysregulated Immune Regulation in the Elderly. FRONTIERS IN AGING 2022; 3:840827. [PMID: 35821823 PMCID: PMC9261323 DOI: 10.3389/fragi.2022.840827] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/30/2022] [Indexed: 12/22/2022]
Abstract
An optimal immune response requires the appropriate interaction between the innate and the adaptive arms of the immune system as well as a proper balance of activation and regulation. After decades of life, the aging immune system is continuously exposed to immune stressors and inflammatory assaults that lead to immune senescence. In this review, we will discuss inflammaging in the elderly, specifically concentrating on IL-6 and IL-1b in the context of T lymphocytes, and how inflammation is related to mortality and morbidities, specifically cardiovascular disease and cancer. Although a number of studies suggests that the anti-inflammatory cytokine TGF-b is elevated in the elderly, heightened inflammation persists. Thus, the regulation of the immune response and the ability to return the immune system to homeostasis is also important. Therefore, we will discuss cellular alterations in aging, concentrating on senescent T cells and CD4+ CD25+ FOXP3+ regulatory T cells (Tregs) in aging
Collapse
Affiliation(s)
- Carey Shive
- Louis Stokes Cleveland VA Medical Center, United States Department of Veterans Affairs, Cleveland, OH, United States.,Case Western Reserve University, Cleveland, OH, United States
| | - Pushpa Pandiyan
- Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
46
|
The role of IL-6 in TBI and PTSD, a potential therapeutic target? Clin Neurol Neurosurg 2022; 218:107280. [PMID: 35567833 DOI: 10.1016/j.clineuro.2022.107280] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/15/2022] [Accepted: 05/02/2022] [Indexed: 01/14/2023]
Abstract
This literature review focuses on the role of IL-6 in TBI or PTSD-induced neuroinflammation. While TBI and PTSD are widely prevalent, these diagnoses are particularly common amongst veterans. Given the role of IL-6 in neuroprotection acutely, compared to detrimental chronically, targeting this cytokine at specific time points may be beneficial in modulating neuroinflammation. Current treatments for TBI or PTSD are variably affective. By reviewing the role of IL-6 in these two diagnoses, future studies can focus on therapeutics to treat neuroinflammation and ultimately reduce the devastating impacts of neuroinflammation on cognition in PTSD and TBI.
Collapse
|
47
|
Wang L, Mohanasundaram P, Lindström M, Asghar MN, Sultana G, Misiorek JO, Jiu Y, Chen H, Chen Z, Toivola DM, Cheng F, Eriksson JE. Vimentin Suppresses Inflammation and Tumorigenesis in the Mouse Intestine. Front Cell Dev Biol 2022; 10:862237. [PMID: 35399505 PMCID: PMC8993042 DOI: 10.3389/fcell.2022.862237] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/22/2022] [Indexed: 01/03/2023] Open
Abstract
Vimentin has been implicated in wound healing, inflammation, and cancer, but its functional contribution to intestinal diseases is poorly understood. To study how vimentin is involved during tissue injury and repair of simple epithelium, we induced colonic epithelial cell damage in the vimentin null (Vim−/−) mouse model. Vim−/− mice challenged with dextran sodium sulfate (DSS) had worse colitis manifestations than wild-type (WT) mice. Vim−/− colons also produced more reactive oxygen and nitrogen species, possibly contributing to the pathogenesis of gut inflammation and tumorigenesis than in WT mice. We subsequently describe that CD11b+ macrophages served as the mainly cellular source of reactive oxygen species (ROS) production via vimentin-ROS-pSTAT3–interleukin-6 inflammatory pathways. Further, we demonstrated that Vim−/− mice did not develop colitis-associated cancer model upon DSS treatment spontaneously but increased tumor numbers and size in the distal colon in the azoxymethane/DSS model comparing with WT mice. Thus, vimentin has a crucial role in protection from colitis induction and tumorigenesis of the colon.
Collapse
Affiliation(s)
- Linglu Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ponnuswamy Mohanasundaram
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Michelle Lindström
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Muhammad Nadeem Asghar
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Giulia Sultana
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Julia O Misiorek
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Department of Molecular Neurooncology, Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznan, Poland
| | - Yaming Jiu
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Hongbo Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Zhi Chen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Diana M Toivola
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| | - Fang Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - John E Eriksson
- Cell Biology, Biosciences, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland.,InFLAMES Research Flagship Center, Åbo Akademi University, Turku, Finland
| |
Collapse
|
48
|
CD4 + T-cell-derived IL-10 promotes CNS inflammation in mice by sustaining effector T cell survival. Cell Rep 2022; 38:110565. [PMID: 35354043 DOI: 10.1016/j.celrep.2022.110565] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 11/24/2022] Open
Abstract
Interleukin (IL)-10 is considered a prototypical anti-inflammatory cytokine, significantly contributing to the maintenance and reestablishment of immune homeostasis. Accordingly, it has been shown in the intestine that IL-10 produced by Tregs can act on effector T cells, thereby limiting inflammation. Herein, we investigate whether this role also applies to IL-10 produced by T cells during central nervous system (CNS) inflammation. During neuroinflammation, both CNS-resident and -infiltrating cells produce IL-10; yet, as IL-10 has a pleotropic function, the exact contribution of the different cellular sources is not fully understood. We find that T-cell-derived IL-10, but not other relevant IL-10 sources, can promote inflammation in experimental autoimmune encephalomyelitis. Furthermore, in the CNS, T-cell-derived IL-10 acts on effector T cells, promoting their survival and thereby enhancing inflammation and CNS autoimmunity. Our data indicate a pro-inflammatory role of T-cell-derived IL-10 in the CNS.
Collapse
|
49
|
Li J, Chen D, Shen M. Tumor Microenvironment Shapes Colorectal Cancer Progression, Metastasis, and Treatment Responses. Front Med (Lausanne) 2022; 9:869010. [PMID: 35402443 PMCID: PMC8984105 DOI: 10.3389/fmed.2022.869010] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/24/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most devastating diseases that accounts for numerous deaths worldwide. Tumor cell-autonomous pathways, such as the oncogenic signaling activation, significantly contribute to CRC progression and metastasis. Recent accumulating evidence suggests that the CRC microenvironment also profoundly promotes or represses this process. As the roles of the tumor microenvironment (TME) in CRC progression and metastasis is gradually uncovered, the importance of these non-cell-autonomous signaling pathways is appreciated. However, we are still at the beginning of this TME function exploring process. In this review, we summarize the current understanding of the TME in CRC progression and metastasis by focusing on the gut microbiota and host cellular and non-cellular components. We also briefly discuss TME-remodeling therapies in CRC.
Collapse
Affiliation(s)
- Jun Li
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Dawei Chen
- Wayne State University School of Medicine, Detroit, MI, United States
| | - Minhong Shen
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Oncology, Wayne State University School of Medicine and Tumor Biology and Microenvironment Research Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, United States
- *Correspondence: Minhong Shen,
| |
Collapse
|
50
|
Kim B, Lee K, Park B. Minecoside promotes apoptotic progression through STAT3 inactivation in breast cancer cells. Oncol Lett 2022; 23:94. [PMID: 35154425 PMCID: PMC8822415 DOI: 10.3892/ol.2022.13214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022] Open
Abstract
Breast cancer is one of the most common malignant tumors in women worldwide, and is a major cause of mortality and morbidity in cancer patients. Constitutive activation of STAT3 has been found in a variety of malignant tumors, including breast cancer. Since STAT3 activation is capable of regulating various important features of tumor cells, identification of a novel STAT3 inhibitor is considered a potential strategy for treating breast cancer. The aim of the present study was to examine whether minecoside (MIN), an active compound extracted from Veronica peregrina L., exerts an antitumor effect by inhibiting STAT3 signaling pathway in MDA-MB-231 cells. The results revealed that MIN inhibited the constitutive STAT3 activation in a dose- and time-dependent manner. MIN also blocked the nuclear translocation of STAT3 and suppressed STAT3-DNA binding. In addition, MIN downregulated the STAT3-mediated expression of proteins such as Bcl-xL, Bcl-2, CXCR4, VEGF, and cyclin D1. Subsequently, MIN promoted the caspase-dependent apoptosis in MDA-MB-231 cells. Overall, results of the present study provide evidence that MIN exerted anticancer activity via inhibition of the STAT3 signaling pathway. Further studies using animal models are required to determine the potential of this molecule as an anticancer drug.
Collapse
Affiliation(s)
- Buyun Kim
- College of Pharmacy, Keimyung University, Dalseo‑Gu, Daegu, North Gyeongsang 704‑701, Republic of Korea
| | - Ki Lee
- College of Pharmacy, Korea University, Sejong 339‑770, Republic of Korea
| | - Byoungduck Park
- College of Pharmacy, Keimyung University, Dalseo‑Gu, Daegu, North Gyeongsang 704‑701, Republic of Korea
| |
Collapse
|