101
|
Hou J, Karin M, Sun B. Targeting cancer-promoting inflammation - have anti-inflammatory therapies come of age? Nat Rev Clin Oncol 2021; 18:261-279. [PMID: 33469195 DOI: 10.1038/s41571-020-00459-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2020] [Indexed: 02/07/2023]
Abstract
The immune system has crucial roles in cancer development and treatment. Whereas adaptive immunity can prevent or constrain cancer through immunosurveillance, innate immunity and inflammation often promote tumorigenesis and malignant progression of nascent cancer. The past decade has witnessed the translation of knowledge derived from preclinical studies of antitumour immunity into clinically effective, approved immunotherapies for cancer. By contrast, the successful implementation of treatments that target cancer-associated inflammation is still awaited. Anti-inflammatory agents have the potential to not only prevent or delay cancer onset but also to improve the efficacy of conventional therapeutics and next-generation immunotherapies. Herein, we review the current clinical advances and experimental findings supporting the utility of an anti-inflammatory approach to the treatment of solid malignancies. Gaining a better mechanistic understanding of the mode of action of anti-inflammatory agents and designing more effective treatment combinations would advance the clinical application of this therapeutic approach.
Collapse
Affiliation(s)
- Jiajie Hou
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Department of Liver Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, University of California San Diego School of Medicine, La Jolla, CA, USA.
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
102
|
Aggen DH, Ager CR, Obradovic A, Chowdhury N, Ghasemzadeh A, Mao W, Chaimowitz M, Lopez-Bujanda ZA, Spina CS, Hawley JE, Dallos MC, Zhang C, Wang V, Li H, Guo X, Drake CG. Blocking IL1 Beta Promotes Tumor Regression and Remodeling of the Myeloid Compartment in a Renal Cell Carcinoma Model: Multidimensional Analyses. Clin Cancer Res 2021; 27:608-621. [PMID: 33148676 PMCID: PMC7980495 DOI: 10.1158/1078-0432.ccr-20-1610] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/11/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Intratumoral immunosuppression mediated by myeloid-derived suppressor cells (MDSC) and tumor-associated macrophages (TAM) represents a potential mechanism of immune checkpoint inhibitor (ICI) resistance in solid tumors. By promoting TAM and MDSC infiltration, IL1β may drive adaptive and innate immune resistance in renal cell carcinoma (RCC) and in other tumor types. EXPERIMENTAL DESIGN Using the RENCA model of RCC, we evaluated clinically relevant combinations of anti-IL1β plus either anti-PD-1 or the multitargeted tyrosine kinase inhibitor (TKI), cabozantinib. We performed comprehensive immune profiling of established RENCA tumors via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA sequencing (RNA-seq). Similar analyses were extended to the MC38 tumor model. RESULTS Analyses via multiparameter flow cytometry, tumor cytokine profiling, and single-cell RNA-seq showed that anti-IL1β reduces infiltration of polymorphonuclear MDSCs and TAMs. Combination treatment with anti-IL1β plus anti-PD-1 or cabozantinib showed increased antitumor activity that was associated with decreases in immunosuppressive MDSCs and increases in M1-like TAMs. CONCLUSIONS Single-cell RNA-seq analyses show that IL1β blockade and ICI or TKI remodel the myeloid compartment through nonredundant, relatively T-cell-independent mechanisms. IL1β is an upstream mediator of adaptive myeloid resistance and represents a potential target for kidney cancer immunotherapy.
Collapse
Affiliation(s)
- David H. Aggen
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Current Address: Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY USA 10065
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Casey R. Ager
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Aleksandar Obradovic
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Nivedita Chowdhury
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Ali Ghasemzadeh
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Wendy Mao
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Current Address: Kite Pharma, 930 Clopper Rd Suite 200, Gaithersburg, MD USA 20878
| | - Matthew Chaimowitz
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Zoila A. Lopez-Bujanda
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA 21205
| | - Catherine S. Spina
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Jessica E. Hawley
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Matthew C. Dallos
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN USA 55905
| | - Vinson Wang
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Hu Li
- Department of Radiation Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Xinzheng Guo
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
| | - Charles G. Drake
- Columbia Center for Translational Immunology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Hematology Oncology, Columbia University Irving Medical Center, New York, NY USA 10032
- Department of Urology, Columbia University Irving Medical Center, New York, NY USA 10032
| |
Collapse
|
103
|
Association between Inflammation and Function of Cell Adhesion Molecules Influence on Gastrointestinal Cancer Development. Cells 2021; 10:cells10010067. [PMID: 33406733 PMCID: PMC7824562 DOI: 10.3390/cells10010067] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/09/2020] [Accepted: 12/29/2020] [Indexed: 12/16/2022] Open
Abstract
Gastrointestinal cancer is highly associated with inflammatory processes inducing the release of cytokines from cancer or immune cells, including interferons, interleukins, chemokines, colony-stimulating factors, and growth factors, which promote or suppress tumor progression. Inflammatory cytokines within the tumor microenvironment promote immune cell infiltration. Infiltrating immune, and tumor-surrounding stromal cells support tumor growth, angiogenesis, metastasis, and immunosuppression through communication with inflammatory cytokines and cell adhesion molecules. Notably, infiltrating immune and tumor cells present immunosuppressive molecules, such as programmed death-ligand 1 (PD-L1) and CD80/CD86. Suppression of cytotoxic T cells promotes tumor avoidance of immune surveillance and greater malignancy. Moreover, glycosylation and sialylation of proteins hyperexpressed on the cancer cell surface have been shown to enhance immune escape and metastasis. Cytokine treatments and immune checkpoint inhibitors are widely used in clinical practice. However, the tumor microenvironment is a rapidly changing milieu involving several factors. In this review, we have provided a summary of the interactions of inflammation and cell adhesion molecules between cancer and other cell types, to improve understanding of the tumor microenvironment.
Collapse
|
104
|
Pyrillou K, Burzynski LC, Clarke MCH. Alternative Pathways of IL-1 Activation, and Its Role in Health and Disease. Front Immunol 2020; 11:613170. [PMID: 33391283 PMCID: PMC7775495 DOI: 10.3389/fimmu.2020.613170] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Cytokines activate or inhibit immune cell behavior and are thus integral to all immune responses. IL-1α and IL-1β are powerful apical cytokines that instigate multiple downstream processes to affect both innate and adaptive immunity. Multiple studies show that IL-1β is typically activated in macrophages after inflammasome sensing of infection or danger, leading to caspase-1 processing of IL-1β and its release. However, many alternative mechanisms activate IL-1α and IL-1β in atypical cell types, and IL-1 function is also important for homeostatic processes that maintain a physiological state. This review focuses on the less studied, yet arguably more interesting biology of IL-1. We detail the production by, and effects of IL-1 on specific innate and adaptive immune cells, report how IL-1 is required for barrier function at multiple sites, and discuss how perturbation of IL-1 pathways can drive disease. Thus, although IL-1 is primarily studied for driving inflammation after release from macrophages, it is clear that it has a multifaceted role that extends far beyond this, with various unconventional effects of IL-1 vital for health. However, much is still unknown, and a detailed understanding of cell-type and context-dependent actions of IL-1 is required to truly understand this enigmatic cytokine, and safely deploy therapeutics for the betterment of human health.
Collapse
Affiliation(s)
| | | | - Murray C. H. Clarke
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
105
|
Kang X, Liang H, Luo Y, Li Z, He F, Han X, Zhang L. Anti-adipogenesis and metabolism-regulating effects of heat-inactivated Streptococcus thermophilus MN-ZLW-002. Lett Appl Microbiol 2020; 72:677-687. [PMID: 32981107 DOI: 10.1111/lam.13398] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/27/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome and obesity have become serious threats to public health worldwide. This study was conducted to evaluate the anti-adipogenesis and metabolism-regulating effects of heat-inactivated Streptococcus thermophilus MN-ZLW-002 (MN-ZLW-002), which can be used as a yogurt starter. In vitro study suggested that MN-ZLW-002 stimulated the RAW264.7 macrophages to produce significant amounts of interleukin (IL)-6, IL-10 and tumour necrosis factor (TNF)-α and induced intense phosphorylation of P38, p44/42 MAPK and nuclear factor κB. MN-ZLW-002-stimulated RAW264.7-conditioned medium (CM) notably suppressed the differentiation and adipogenesis of 3T3-L1 pre-adipocytes. The 12-week in vivo study suggested that orally administered MN-ZLW-002 significantly reduced the weight gain of mice caused by the high-fat diet (HFD) at weeks 3-8; decreased fasting blood glucose levels at week 4 and week 8; decreased serum total triglyceride level at week 12. MN-ZLW-002 also reduced serum IL-1β and chemokine ligand 3 levels in the HFD-fed mice. These findings suggest that heat-inactivated MN-ZLW-002 can suppress adipocytes differentiation and lipid accumulation by regulating the immune response, possibly via the release of cytokines, particularly TNF-α; MN-ZLW-002 can improve metabolism-related indicators in the early stage of HFD intervention and regulate the related pro-inflammatory immune response.
Collapse
Affiliation(s)
- X Kang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - H Liang
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Y Luo
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Z Li
- R&D Center, Inner Mongolia Meng Niu Dairy Industry (Group) Co., Ltd, Hohhot, Inner Mongolia, China
| | - F He
- Department of Nutrition, Food Safety and Toxicology, West China School of Public Health and West China Forth Hospital, Sichuan University, Chengdu, Sichuan, China
| | - X Han
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - L Zhang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang, China.,College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| |
Collapse
|
106
|
Hu F, Guo F, Zhu Y, Zhou Q, Li T, Xiang H, Shang D. IL-17 in pancreatic disease: pathogenesis and pharmacotherapy. Am J Cancer Res 2020; 10:3551-3564. [PMID: 33294254 PMCID: PMC7716161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/14/2020] [Indexed: 06/12/2023] Open
Abstract
Increasing evidence highlights the role of the interleukin (IL)-17 family in pancreatic diseases. IL-17A induces acinar cell injury directly, recruits neutrophils, and cooperates with other inflammatory factors to exacerbate pancreatic inflammation. It also triggers islet β-cell apoptosis and nitric oxide-dependent cytotoxicity, thus aggravating islet inflammation. IL-17A seems to have different roles in pancreatic intraepithelial neoplasia (PanIN) and pancreatic cancer (PC). IL-17A participates in the progression of acinar-ductal metaplasia (ADM) and PanIN, but not related to the characteristics of PC stem cells and the overall survival of patients. Acting similar to IL-17A, IL-17B accelerates the invasion and metastasis of PC, and predicts prognosis of PC and the therapeutic effect of gemcitabine. Herein, we review the current understanding of the pathogenesis of IL-17 in pancreatitis, type 1 diabetes mellitus (T1DM), and PC, as well as potential pharmacotherapy targeting IL-17 and its receptors in pancreatic diseases. The findings summarized in this article are of considerable significance for understanding the essential role of IL-17 in pancreatic diseases.
Collapse
Affiliation(s)
- Fenglin Hu
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
- Department of General Surgery, Pancreatic-Biliary Center, First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Fangyue Guo
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Yutong Zhu
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Qi Zhou
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Tongming Li
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Hong Xiang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, China
| | - Dong Shang
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical UniversityDalian 116000, Liaoning, China
- Department of General Surgery, Pancreatic-Biliary Center, First Affiliated Hospital of Dalian Medical UniversityDalian 116000, Liaoning, China
| |
Collapse
|
107
|
Zhang Y, Dong X, Bai L, Shang X, Zeng Y. MUC1-induced immunosuppression in colon cancer can be reversed by blocking the PD1/PDL1 signaling pathway. Oncol Lett 2020; 20:317. [PMID: 33133253 PMCID: PMC7590440 DOI: 10.3892/ol.2020.12180] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 08/20/2020] [Indexed: 12/14/2022] Open
Abstract
Mucin1 (MUC1) upregulation in colon cancer has been linked to poor patient outcomes and advanced stage at diagnosis. This is partially due to MUC1-mediated inhibition of T-cell proliferation affecting efficient lysis by cytotoxic lymphocytes, which contributes to escape from immune surveillance. In the present study, human colorectal cancer tissues were collected, and MUC1-positive and MUC1-negative colon cancer mouse models were prepared; subsequently, the number and function of immune cells in tumor tissues were measured using flow cytometry. The present study revealed that MUC1, as a tumor-associated antigen, can recruit more tumor-infiltrating lymphocytes into the tumor microenvironment compared with MUC1-negative colon cancer, but that these cells could not serve antitumor roles. Conversely, the present study demonstrated that MUC1-positive colon cancer attracted more regulatory T cells (Treg cells), myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) to the tumor site than MUC1-negative colon cancer. Furthermore, the data suggested that programmed death protein 1 (PD1)-programmed death ligand 1 (PDL1) expression is greater in MUC1-positive colon cancer. Blocking the PD1-PDL1 signaling pathway reduced the percentage of Treg cells, MDSCs and TAMs in the tumor microenvironment, enhanced T-cell cytotoxicity and inhibited tumor growth, prolonging the survival time of MUC1-positive tumor-bearing mice. Therefore, the present study elucidated the role of MUC1 in tumor immune escape and provides a foundation for the application of PDL1 inhibitors to MUC1-positive colon cancer.
Collapse
Affiliation(s)
- Yinghui Zhang
- Department of Gastroenterology, The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Xiangqian Dong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Institute of Digestive Diseases, Kunming, Yunnan 650032, P.R. China
| | - Liping Bai
- Department of Gastroenterology, The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Xueqin Shang
- Department of Medical Oncology, The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650021, P.R. China
| | - Yujian Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
108
|
Janney A, Powrie F, Mann EH. Host–microbiota maladaptation in colorectal cancer. Nature 2020; 585:509-517. [DOI: 10.1038/s41586-020-2729-3] [Citation(s) in RCA: 272] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022]
|
109
|
The Macrophages-Microbiota Interplay in Colorectal Cancer (CRC)-Related Inflammation: Prognostic and Therapeutic Significance. Int J Mol Sci 2020; 21:ijms21186866. [PMID: 32962159 PMCID: PMC7558485 DOI: 10.3390/ijms21186866] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor-associated macrophages (TAMs) are the main population of myeloid cells infiltrating solid tumors and the pivotal orchestrators of cancer-promoting inflammation. However, due to their exceptional plasticity, macrophages can be also key effector cells and powerful activators of adaptive anti-tumor immunity. This functional heterogeneity is emerging in human tumors, colorectal cancer (CRC) in particular, where the dynamic co-existence of different macrophage subtypes influences tumor development, outcome, and response to therapies. Intestinal macrophages are in close interaction with enteric microbiota, which contributes to carcinogenesis and affects treatment outcomes. This interplay may be particularly relevant in CRC, one of the most prevalent and lethal cancer types in the world. Therefore, both macrophages and intestinal microbiota are considered promising prognostic indicators and valuable targets for new therapeutic approaches. Here, we discuss the current understanding of the molecular circuits underlying the interplay between macrophages and microbiota in CRC development, progression, and response to both conventional therapies and immunotherapies.
Collapse
|
110
|
Jaillon S, Ponzetta A, Di Mitri D, Santoni A, Bonecchi R, Mantovani A. Neutrophil diversity and plasticity in tumour progression and therapy. Nat Rev Cancer 2020; 20:485-503. [PMID: 32694624 DOI: 10.1038/s41568-020-0281-y] [Citation(s) in RCA: 716] [Impact Index Per Article: 143.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/11/2022]
Abstract
Neutrophils play a key role in defence against infection and in the activation and regulation of innate and adaptive immunity. In cancer, tumour-associated neutrophils (TANs) have emerged as an important component of the tumour microenvironment. Here, they can exert dual functions. TANs can be part of tumour-promoting inflammation by driving angiogenesis, extracellular matrix remodelling, metastasis and immunosuppression. Conversely, neutrophils can also mediate antitumour responses by direct killing of tumour cells and by participating in cellular networks that mediate antitumour resistance. Neutrophil diversity and plasticity underlie the dual potential of TANs in the tumour microenvironment. Myeloid checkpoints as well as the tumour and tissue contexture shape neutrophil function in response to conventional therapies and immunotherapy. We surmise that neutrophils can provide tools to tailor current immunotherapy strategies and pave the way to myeloid cell-centred therapeutic strategies, which would be complementary to current approaches.
Collapse
Affiliation(s)
- Sebastien Jaillon
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
| | - Andrea Ponzetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Diletta Di Mitri
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Angela Santoni
- Dipartimento di Medicina Molecolare Istituto Pasteur-Fondazione Cenci Bolognetti, Università di Roma 'La Sapienza', Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Raffaella Bonecchi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy.
- Humanitas Clinical and Research Center IRCCS, Rozzano (MI), Italy.
- The William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
111
|
Rébé C, Ghiringhelli F. Interleukin-1β and Cancer. Cancers (Basel) 2020; 12:E1791. [PMID: 32635472 PMCID: PMC7408158 DOI: 10.3390/cancers12071791] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Within a tumor, IL-1β is produced and secreted by various cell types, such as immune cells, fibroblasts, or cancer cells. The IL1B gene is induced after "priming" of the cells and a second signal is required to allow IL-1β maturation by inflammasome-activated caspase-1. IL-1β is then released and leads to transcription of target genes through its ligation with IL-1R1 on target cells. IL-1β expression and maturation are guided by gene polymorphisms and by the cellular context. In cancer, IL-1β has pleiotropic effects on immune cells, angiogenesis, cancer cell proliferation, migration, and metastasis. Moreover, anti-cancer treatments are able to promote IL-1β production by cancer or immune cells, with opposite effects on cancer progression. This raises the question of whether or not to use IL-1β inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Cédric Rébé
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Centre Georges François Leclerc, INSERM LNC UMR1231, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| |
Collapse
|
112
|
Pinteaux E, Abdulaal WH, Mufazalov IA, Humphreys NE, Simonsen-Jackson M, Francis S, Müller W, Waisman A. Cell-specific conditional deletion of interleukin-1 (IL-1) ligands and its receptors: a new toolbox to study the role of IL-1 in health and disease. J Mol Med (Berl) 2020; 98:923-930. [PMID: 32468079 PMCID: PMC7343756 DOI: 10.1007/s00109-020-01928-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 01/08/2023]
Abstract
The pro-inflammatory cytokine interleukin-1 (IL-1) plays a key role in many physiological processes and during the inflammatory and immune response to most common diseases. IL-1 exists as two agonists, IL-1α and IL-1β that bind to the only signaling IL-1 type 1 receptor (IL-1R1), while a second decoy IL-1 type 2 receptor (IL-1R2) binds both forms of IL-1 without inducing cell signaling. The field of immunology and inflammation research has, over the past 35 years, unraveled many mechanisms of IL-1 actions, through in vitro manipulation of the IL-1 system or by using genetically engineered mouse models that lack either member of the IL-1 family in ubiquitous constitutive manner. However, the limitation of global mouse knockout technology has significantly hampered our understanding of the precise mechanisms of IL-1 actions in animal models of disease. Here we report and review the recent generation of new conditional mouse mutants in which exons of Il1a, Il1b, Il1r1, and Il1r2 genes flanked by loxP sites (fl/fl) can be deleted in cell-/tissue-specific constitutive or inducible manner by Cre recombinase expression. Hence, IL-1αfl/fl, IL-1βfl/fl, IL-1R1fl/fl, and IL-1R2fl/fl mice constitute a new toolbox that will provide a step change in our understanding of the cell-specific role of IL-1 and its receptor in health and disease and the potential development of targeted IL-1 therapies.
Collapse
Affiliation(s)
- Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom.
| | - Wesam H Abdulaal
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, P.O.BOX 80203, Jeddah, 21589, Kingdom of Saudi Arabia
| | - Ilgiz A Mufazalov
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Langenbeckstrasse 1, Building 308A, 55131, Mainz, Germany
| | - Neil E Humphreys
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
- Epigenetics and Neurobiology Unit, Adriano Buzzati-Traverso Campus, EMBL-Rome, Via Ramarini, 3200015, Monterotondo, RM, Italy
| | - Maj Simonsen-Jackson
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Sheila Francis
- Department of Infection, Immunity & Cardiovascular Disease, Medical School, University of Sheffield, S10 2RX, Sheffield, United Kingdom
| | - Werner Müller
- Faculty of Biology, Medicine and Health, University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, United Kingdom
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University of Mainz, Langenbeckstrasse 1, Building 308A, 55131, Mainz, Germany
| |
Collapse
|
113
|
Wei HX, Wang B, Li B. IL-10 and IL-22 in Mucosal Immunity: Driving Protection and Pathology. Front Immunol 2020; 11:1315. [PMID: 32670290 PMCID: PMC7332769 DOI: 10.3389/fimmu.2020.01315] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022] Open
Abstract
The barrier surfaces of the gastrointestinal tract are in constant contact with various microorganisms. Cytokines orchestrate the mucosal adaptive and innate immune cells in the defense against pathogens. IL-10 and IL-22 are the best studied members of the IL-10 family and play essential roles in maintaining mucosal homeostasis. IL-10 serves as an important regulator in preventing pro-inflammatory responses while IL-22 plays a protective role in tissue damage and contributes to pathology in certain settings. In this review, we focus on these two cytokines in the development of gastrointestinal diseases, including inflammatory bowel diseases (IBD) and colitis-associated cancer (CAC). We summarize the recent studies and try to gain a better understanding on how they regulate immune responses to maintain equilibrium under inflammatory conditions.
Collapse
Affiliation(s)
- Hua-Xing Wei
- Division of Life Sciences and Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Baolong Wang
- Division of Life Sciences and Medicine, Department of Laboratory Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Bofeng Li
- Division of Life Sciences and Medicine, Department of Medical Oncology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| |
Collapse
|
114
|
Loft ND, Vaengebjerg S, Skov L. Cancer risk in patients with psoriasis: should we be paying more attention? Expert Rev Clin Immunol 2020; 16:479-492. [DOI: 10.1080/1744666x.2020.1754194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Nikolai Dyrberg Loft
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Sofie Vaengebjerg
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, Copenhagen, Denmark
| | - Lone Skov
- Department of Dermatology and Allergy, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Research Group for Inflammatory Skin, Herlev and Gentofte Hospital, Copenhagen, Denmark
| |
Collapse
|
115
|
The complexity of neutrophils in health and disease: Focus on cancer. Semin Immunol 2020; 48:101409. [PMID: 32958359 PMCID: PMC7500440 DOI: 10.1016/j.smim.2020.101409] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/14/2022]
Abstract
Neutrophils are essential soldiers of the immune response and their role have long been restricted to their activities in defence against microbial infections and during the acute phase of the inflammatory response. However, increasing number of investigations showed that neutrophils are endowed with plasticity and can participate in the orchestration of both innate and adaptive immune responses. Neutrophils have an impact on a broad range of disorders, including infections, chronic inflammations, and cancer. Neutrophils are present in the tumour microenvironment and have been reported to mediate both pro-tumour and anti-tumour responses. Neutrophils can contribute to genetic instability, tumour cell proliferation, angiogenesis and suppression of the anti-tumour immune response. In contrast, neutrophils are reported to mediate anti-tumour resistance by direct killing of tumour cells or by engaging cooperative interactions with other immune cells. Here we discuss the current understandings of neutrophils biology and functions in health and diseases, with a specific focus on their role in cancer biology and their prognostic significance in human cancer.
Collapse
|
116
|
Immunotherapy, Inflammation and Colorectal Cancer. Cells 2020; 9:cells9030618. [PMID: 32143413 PMCID: PMC7140520 DOI: 10.3390/cells9030618] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/29/2020] [Accepted: 03/03/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer type, and third highest in mortality rates among cancer-related deaths in the United States. Originating from intestinal epithelial cells in the colon and rectum, that are impacted by numerous factors including genetics, environment and chronic, lingering inflammation, CRC can be a problematic malignancy to treat when detected at advanced stages. Chemotherapeutic agents serve as the historical first line of defense in the treatment of metastatic CRC. In recent years, however, combinational treatment with targeted therapies, such as vascular endothelial growth factor, or epidermal growth factor receptor inhibitors, has proven to be quite effective in patients with specific CRC subtypes. While scientific and clinical advances have uncovered promising new treatment options, the five-year survival rate for metastatic CRC is still low at about 14%. Current research into the efficacy of immunotherapy, particularly immune checkpoint inhibitor therapy (ICI) in mismatch repair deficient and microsatellite instability high (dMMR-MSI-H) CRC tumors have shown promising results, but its use in other CRC subtypes has been either unsuccessful, or not extensively explored. This Review will focus on the current status of immunotherapies, including ICI, vaccination and adoptive T cell therapy (ATC) in the treatment of CRC and its potential use, not only in dMMR-MSI-H CRC, but also in mismatch repair proficient and microsatellite instability low (pMMR-MSI-L).
Collapse
|
117
|
Keir ME, Yi T, Lu TT, Ghilardi N. The role of IL-22 in intestinal health and disease. J Exp Med 2020; 217:e20192195. [PMID: 32997932 PMCID: PMC7062536 DOI: 10.1084/jem.20192195] [Citation(s) in RCA: 290] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/24/2022] Open
Abstract
The cytokine interleukin-22 (IL-22) is a critical regulator of epithelial homeostasis. It has been implicated in multiple aspects of epithelial barrier function, including regulation of epithelial cell growth and permeability, production of mucus and antimicrobial proteins (AMPs), and complement production. In this review, we focus specifically on the role of IL-22 in the intestinal epithelium. We summarize recent advances in our understanding of how IL-22 regulates homeostasis and host defense, and we discuss the IL-22 pathway as a therapeutic target in diseases of the intestine, including inflammatory bowel disease (IBD), graft-versus-host disease (GVHD), and cancer.
Collapse
Affiliation(s)
- Mary E. Keir
- Biomarker Discovery, Genentech, South San Francisco, CA
| | - Tangsheng Yi
- Department of Immunology, Genentech, South San Francisco, CA
| | - Timothy T. Lu
- Early Clinical Development, Genentech, South San Francisco, CA
| | | |
Collapse
|
118
|
Shalapour S, Karin M. Cruel to Be Kind: Epithelial, Microbial, and Immune Cell Interactions in Gastrointestinal Cancers. Annu Rev Immunol 2020; 38:649-671. [PMID: 32040356 DOI: 10.1146/annurev-immunol-082019-081656] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A plethora of experimental and epidemiological evidence supports a critical role for inflammation and adaptive immunity in the onset of cancer and in shaping its response to therapy. These data are particularly robust for gastrointestinal (GI) cancers, such as those affecting the GI tract, liver, and pancreas, on which this review is focused. We propose a unifying hypothesis according to which intestinal barrier disruption is the origin of tumor-promoting inflammation that acts in conjunction with tissue-specific cancer-initiating mutations. The gut microbiota and its products impact tissue-resident and recruited myeloid cells that promote tumorigenesis through secretion of growth- and survival-promoting cytokines that act on epithelial cells, as well as fibrogenic and immunosuppressive cytokines that interfere with the proper function of adaptive antitumor immunity. Understanding these relationships should improve our ability to prevent cancer development and stimulate the immune system to eliminate existing malignancies.
Collapse
Affiliation(s)
- Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA; , .,Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA; , .,Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California 92093, USA.,Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|
119
|
Zhang W, Borcherding N, Kolb R. IL-1 Signaling in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1240:1-23. [PMID: 32060884 DOI: 10.1007/978-3-030-38315-2_1] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Interleukin 1 (IL-1) has long been known for its pleiotropic effects on inflammation that plays a complex, and sometimes contrasting, role in different stages of cancer development. As a major proinflammatory cytokine, IL-1β is mainly expressed by innate immune cells. IL-1α, however, is expressed by various cell types under physiological and pathological conditions. IL-1R1 is the main receptor for both ligands and is expressed by various cell types, including innate and adaptive immune cell types, epithelial cells, endothelial cells, adipocytes, chondrocytes, fibroblasts, etc. IL-1 and IL-1R1 receptor interaction leads to a set of common signaling pathways, mainly the NF-kB and MAP kinase pathways, as a result of complex positive and negative regulations. The variety of cell types with IL-1R1 expression dictates the role of IL-1 signaling at different stages of cancer, which under certain circumstances leads to contrasting roles in tumor development. Recent availability of IL-1R1 conditional knockout mouse model has made it possible to dissect the role of IL-1/IL-1R1 signaling transduction in different cell types within the tumor microenvironment. This chapter will focus on the role of IL-1/IL-1R1 in different cell types within the tumor microenvironment and discuss the potential of targeting this pathway in cancer therapy.
Collapse
Affiliation(s)
- Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, USA.
| | | | - Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, UF Health Cancer Center, University of Florida, Gainesville, FL, USA
| |
Collapse
|
120
|
Shalapour S, Karin M. Pas de Deux: Control of Anti-tumor Immunity by Cancer-Associated Inflammation. Immunity 2019; 51:15-26. [PMID: 31315033 DOI: 10.1016/j.immuni.2019.06.021] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/11/2019] [Accepted: 06/24/2019] [Indexed: 12/14/2022]
Abstract
In many settings, tumor-associated inflammation, supported mainly by innate immune cells, contributes to tumor growth. Initial innate activation triggers secretion of inflammatory, regenerative, and anti-inflammatory cytokines, which in turn shape the adaptive immune response to the tumor. Here, we review the current understanding of the intricate dialog between cancer-associated inflammation and anti-tumor immunity. We discuss the changing nature of these interactions during tumor progression and the impact of the tissue environment on the anti-tumor immune response. In this context, we outline important gaps in current understanding by considering basic research and findings in the clinic. The future of cancer immunotherapy and its utility depend on improved understanding of these interactions and the ability to manipulate them in a predictable and beneficial manner.
Collapse
Affiliation(s)
- Shabnam Shalapour
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Department of Pharmacology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| |
Collapse
|
121
|
Greten FR, Grivennikov SI. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019; 51:27-41. [PMID: 31315034 DOI: 10.1016/j.immuni.2019.06.025] [Citation(s) in RCA: 2358] [Impact Index Per Article: 393.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/21/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
Inflammation predisposes to the development of cancer and promotes all stages of tumorigenesis. Cancer cells, as well as surrounding stromal and inflammatory cells, engage in well-orchestrated reciprocal interactions to form an inflammatory tumor microenvironment (TME). Cells within the TME are highly plastic, continuously changing their phenotypic and functional characteristics. Here, we review the origins of inflammation in tumors, and the mechanisms whereby inflammation drives tumor initiation, growth, progression, and metastasis. We discuss how tumor-promoting inflammation closely resembles inflammatory processes typically found during development, immunity, maintenance of tissue homeostasis, or tissue repair and illuminate the distinctions between tissue-protective and pro-tumorigenic inflammation, including spatiotemporal considerations. Defining the cornerstone rules of engagement governing molecular and cellular mechanisms of tumor-promoting inflammation will be essential for further development of anti-cancer therapies.
Collapse
Affiliation(s)
- Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, 60596 Frankfurt/Main, Germany; Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt/Main, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Sergei I Grivennikov
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, 19111, USA.
| |
Collapse
|
122
|
Myeloid immunosuppression and immune checkpoints in the tumor microenvironment. Cell Mol Immunol 2019; 17:1-12. [PMID: 31611651 DOI: 10.1038/s41423-019-0306-1] [Citation(s) in RCA: 338] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/17/2019] [Indexed: 02/08/2023] Open
Abstract
Tumor-promoting inflammation and the avoidance of immune destruction are hallmarks of cancer. While innate immune cells, such as neutrophils, monocytes, and macrophages, are critical mediators for sterile and nonsterile inflammation, persistent inflammation, such as that which occurs in cancer, is known to disturb normal myelopoiesis. This disturbance leads to the generation of immunosuppressive myeloid cells, such as myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs). Due to their potent suppressive activities against effector lymphocytes and their abundance in the tumor microenvironment, immunosuppressive myeloid cells act as a major barrier to cancer immunotherapy. Indeed, various therapeutic approaches directed toward immunosuppressive myeloid cells are actively being tested in preclinical and clinical studies. These include anti-inflammatory agents, therapeutic blockade of the mobilization and survival of myeloid cells, and immunostimulatory adjuvants. More recently, immune checkpoint molecules expressed on tumor-infiltrating myeloid cells have emerged as potential therapeutic targets to redirect these cells to eliminate tumor cells. In this review, we discuss the complex crosstalk between cancer-related inflammation and immunosuppressive myeloid cells and possible therapeutic strategies to harness antitumor immune responses.
Collapse
|
123
|
Pandey A, Shen C, Man SM. Inflammasomes in Colitis and Colorectal Cancer: Mechanism of Action and Therapies. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2019; 92:481-498. [PMID: 31543710 PMCID: PMC6747943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Colorectal cancer is a multifactorial disease and a leading cause of cancer-related deaths worldwide. Inflammation is a driver across multiple stages in the development of colorectal cancer. The inflammasome is a cytosolic multiprotein complex of the innate immune system central to the regulation of inflammation, pyroptosis, and other cellular processes important for maintaining gut homeostasis. Studies using mouse models of colitis and colitis-associated colorectal cancer have highlighted diverse and sometimes contrasting roles of inflammasomes in maintaining a balance between intestinal barrier function and the gut microbiota. In addition, persistent and/or dysregulated stimulation of inflammasome sensors finetune inflammation and tumorigenesis in the intestine. This review highlights the emerging role of inflammasome signaling in colitis and colitis-associated colorectal cancer. We also review the key mechanisms by which inflammasome signaling modulate inflammation and tumor development. Finally, we speculate the importance of using more tightly regulated experimental approaches to examine the role of gut microbiota in colorectal cancer.
Collapse
Affiliation(s)
| | | | - Si Ming Man
- To whom all correspondence should be addressed: Si Ming Man, Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, 2601, Australia; Tel: 61 2 612 56793,
| |
Collapse
|
124
|
Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol 2019; 20:970-979. [PMID: 31235952 DOI: 10.1038/s41590-019-0415-0] [Citation(s) in RCA: 450] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/01/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis are characterized by uncontrolled activation of intestinal immune cells in a genetically susceptible host. Due to the progressive and destructive nature of the inflammatory process in IBD, complications such as fibrosis, stenosis or cancer are frequently observed, which highlights the need for effective anti-inflammatory therapy. Studies have identified altered trafficking of immune cells and pathogenic immune cell circuits as crucial drivers of mucosal inflammation and tissue destruction in IBD. A defective gut barrier and microbial dysbiosis induce such accumulation and local activation of immune cells, which results in a pro-inflammatory cytokine loop that overrides anti-inflammatory signals and causes chronic intestinal inflammation. This Review discusses pathogenic cytokine responses of immune cells as well as immune cell trafficking as a rational basis for new translational therapies in IBD.
Collapse
|
125
|
Combining use of Phillyrin and autophagy blocker alleviates laryngeal squamous cell carcinoma via AMPK/mTOR/p70S6K signaling. Biosci Rep 2019; 39:BSR20190459. [PMID: 31147451 PMCID: PMC6616054 DOI: 10.1042/bsr20190459] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/07/2019] [Accepted: 05/21/2019] [Indexed: 12/15/2022] Open
Abstract
Phillyrin (PHN), one of the major active constituents of Forsythia suspensa and F. koreana, has been reported to produce antioxidant, antibacterial, anti-obesity and anti-inflammatory effects. However, no study has demonstrated the role of PHN in laryngeal squamous cell carcinoma (LSCC). We aimed to investigate the effects of PHN on the proliferation and apoptosis of HEp-2 cells. In the present study, PHN alone showed little effect on HEp-2 cell proliferation and apoptosis. Subsequent tests showed that PHN could largely enhance the level of autophagy on HEp-2 cells. Combining use of PHN and autophagy blockers including 3-methyladenine (3-MA) and chloroquine (CQ) significantly inhibited HEp-2 cell proliferation in a dose- and time-dependent manner and induced apoptosis after 24 h in a dose-dependent manner. Additionally, we found that the possible underlying molecular mechanism of PHN-induced autophagy might be through the AMPK/mTOR/p70S6K signaling pathway. Taken together, our study indicates that combining use of PHN and autophagy blockers may serve as a novel strategy in LSCC treatment.
Collapse
|
126
|
Baker KJ, Houston A, Brint E. IL-1 Family Members in Cancer; Two Sides to Every Story. Front Immunol 2019; 10:1197. [PMID: 31231372 PMCID: PMC6567883 DOI: 10.3389/fimmu.2019.01197] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/13/2019] [Indexed: 12/22/2022] Open
Abstract
The IL-1 family of cytokines currently comprises of seven ligands with pro-inflammatory activity (IL-1α and IL-1β, IL-18, IL-33, IL-36α, IL-36β, IL-36γ) as well as two ligands with anti-inflammatory activity (IL-37, IL-38). These cytokines are known to play a key role in modulating both the innate and adaptive immunes response, with dysregulation linked to a variety of autoimmune and inflammatory diseases. Given the increasing appreciation of the link between inflammation and cancer, the role of several members of this family in the pathogenesis of cancer has been extensively investigated. In this review, we highlight both the pro- and anti-tumorigenic effects identified for almost all members of this family, and explore potential underlying mechanisms accounting for these divergent effects. Such dual functions need to be carefully assessed when developing therapeutic intervention strategies targeting these cytokines in cancer.
Collapse
Affiliation(s)
- Kevin J Baker
- Department of Pathology, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland.,APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Aileen Houston
- Department of Medicine, University College Cork, Cork, Ireland.,CancerResearch@UCC, University College Cork, Cork, Ireland
| | - Elizabeth Brint
- Department of Pathology, University College Cork, Cork, Ireland.,CancerResearch@UCC, University College Cork, Cork, Ireland
| |
Collapse
|
127
|
Cataisson C, Salcedo R, Michalowski AM, Klosterman M, Naik S, Li L, Pan MJ, Sweet A, Chen JQ, Kostecka LG, Karwan M, Smith L, Dai RM, Stewart CA, Lyakh L, Hsieh WT, Khan A, Yang H, Lee M, Trinchieri G, Yuspa SH. T-Cell Deletion of MyD88 Connects IL17 and IκBζ to RAS Oncogenesis. Mol Cancer Res 2019; 17:1759-1773. [PMID: 31164412 DOI: 10.1158/1541-7786.mcr-19-0227] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 05/13/2019] [Accepted: 05/30/2019] [Indexed: 01/15/2023]
Abstract
Cancer development requires a favorable tissue microenvironment. By deleting Myd88 in keratinocytes or specific bone marrow subpopulations in oncogenic RAS-mediated skin carcinogenesis, we show that IL17 from infiltrating T cells and IκBζ signaling in keratinocytes are essential to produce a permissive microenvironment and tumor formation. Both normal and RAS-transformed keratinocytes respond to tumor promoters by activating canonical NF-κB and IκBζ signaling, releasing specific cytokines and chemokines that attract Th17 cells through MyD88-dependent signaling in T cells. The release of IL17 into the microenvironment elevates IκBζ in normal and RAS-transformed keratinocytes. Activation of IκBζ signaling is required for the expression of specific promoting factors induced by IL17 in normal keratinocytes and constitutively expressed in RAS-initiated keratinocytes. Deletion of Nfkbiz in keratinocytes impairs RAS-mediated benign tumor formation. Transcriptional profiling and gene set enrichment analysis of IκBζ-deficient RAS-initiated keratinocytes indicate that IκBζ signaling is common for RAS transformation of multiple epithelial cancers. Probing The Cancer Genome Atlas datasets using this transcriptional profile indicates that reduction of IκBζ signaling during cancer progression associates with poor prognosis in RAS-driven human cancers. IMPLICATIONS: The paradox that elevation of IκBζ and stimulation of IκBζ signaling through tumor extrinsic factors is required for RAS-mediated benign tumor formation while relative IκBζ expression is reduced in advanced cancers with poor prognosis implies that tumor cells switch from microenvironmental dependency early in carcinogenesis to cell-autonomous pathways during cancer progression.
Collapse
Affiliation(s)
| | - Rosalba Salcedo
- Cancer and Inflammation Program (CIP), NCI, Bethesda Maryland
| | | | - Mary Klosterman
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Shruti Naik
- Department of Pathology and Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, New York
| | - Luowei Li
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Michelle J Pan
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Amalia Sweet
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource, Center for Cancer Research, NCI, Bethesda, Maryland
| | | | - Megan Karwan
- Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Loretta Smith
- Cancer and Inflammation Program (CIP), NCI, Bethesda Maryland
| | - Ren-Ming Dai
- Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - Lyudmila Lyakh
- Cancer and Inflammation Program (CIP), NCI, Bethesda Maryland.,Division of Allergy, Immunology & Transplantation, National Institute of Allergy and Infectious Diseases, NIH, Bethesda Maryland
| | | | - Asra Khan
- Cancer and Inflammation Program (CIP), NCI, Bethesda Maryland
| | - Howard Yang
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | - Maxwell Lee
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland
| | | | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, NCI, Bethesda, Maryland.
| |
Collapse
|
128
|
Mantovani A, Dinarello CA, Molgora M, Garlanda C. Interleukin-1 and Related Cytokines in the Regulation of Inflammation and Immunity. Immunity 2019; 50:778-795. [PMID: 30995499 PMCID: PMC7174020 DOI: 10.1016/j.immuni.2019.03.012] [Citation(s) in RCA: 696] [Impact Index Per Article: 116.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/06/2023]
Abstract
Forty years after its naming, interleukin-1 (IL-1) is experiencing a renaissance brought on by the growing understanding of its context-dependent roles and advances in the clinic. Recent studies have identified important roles for members of the IL-1 family-IL-18, IL-33, IL-36, IL-37, and IL-38-in inflammation and immunity. Here, we review the complex functions of IL-1 family members in the orchestration of innate and adaptive immune responses and their diversity and plasticity. We discuss the varied roles of IL-1 family members in immune homeostasis and their contribution to pathologies, including autoimmunity and auto-inflammation, dysmetabolism, cardiovascular disorders, and cancer. The trans-disease therapeutic activity of anti-IL-1 strategies argues for immunity and inflammation as a metanarrative of modern medicine.
Collapse
Affiliation(s)
- Alberto Mantovani
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy; William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | - Charles A Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA; Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Martina Molgora
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy
| | - Cecilia Garlanda
- IRCCS Humanitas Clinical and Research Center, via Manzoni 56, 20089 Rozzano Milan, Italy; Humanitas University, via Rita Levi Montalcini, 20090 Pieve Emanuele Milan, Italy.
| |
Collapse
|
129
|
Abstract
Inflammasomes are molecular platforms that assemble upon sensing various intracellular stimuli. Inflammasome assembly leads to activation of caspase 1, thereby promoting the secretion of bioactive interleukin-1β (IL-1β) and IL-18 and inducing an inflammatory cell death called pyroptosis. Effectors of the inflammasome efficiently drive an immune response, primarily providing protection against microbial infections and mediating control over sterile insults. However, aberrant inflammasome signalling is associated with pathogenesis of inflammatory and metabolic diseases, neurodegeneration and malignancies. Chronic inflammation perpetuated by inflammasome activation plays a central role in all stages of tumorigenesis, including immunosuppression, proliferation, angiogenesis and metastasis. Conversely, inflammasome signalling also contributes to tumour suppression by maintaining intestinal barrier integrity, which portrays the diverse roles of inflammasomes in tumorigenesis. Studies have underscored the importance of environmental factors, such as diet and gut microbiota, in inflammasome signalling, which in turn influences tumorigenesis. In this Review, we deliver an overview of the interplay between inflammasomes and tumorigenesis and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|