101
|
Herrera-Campos AB, Zamudio-Martinez E, Delgado-Bellido D, Fernández-Cortés M, Montuenga LM, Oliver FJ, Garcia-Diaz A. Implications of Hyperoxia over the Tumor Microenvironment: An Overview Highlighting the Importance of the Immune System. Cancers (Basel) 2022; 14:2740. [PMID: 35681719 PMCID: PMC9179641 DOI: 10.3390/cancers14112740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/04/2023] Open
Abstract
Hyperoxia is used in order to counteract hypoxia effects in the TME (tumor microenvironment), which are described to boost the malignant tumor phenotype and poor prognosis. The reduction of tumor hypoxic state through the formation of a non-aberrant vasculature or an increase in the toxicity of the therapeutic agent improves the efficacy of therapies such as chemotherapy. Radiotherapy efficacy has also improved, where apoptotic mechanisms seem to be implicated. Moreover, hyperoxia increases the antitumor immunity through diverse pathways, leading to an immunopermissive TME. Although hyperoxia is an approved treatment for preventing and treating hypoxemia, it has harmful side-effects. Prolonged exposure to high oxygen levels may cause acute lung injury, characterized by an exacerbated immune response, and the destruction of the alveolar-capillary barrier. Furthermore, under this situation, the high concentration of ROS may cause toxicity that will lead not only to cell death but also to an increase in chemoattractant and proinflammatory cytokine secretion. This would end in a lung leukocyte recruitment and, therefore, lung damage. Moreover, unregulated inflammation causes different consequences promoting tumor development and metastasis. This process is known as protumor inflammation, where different cell types and molecules are implicated; for instance, IL-1β has been described as a key cytokine. Although current results show benefits over cancer therapies using hyperoxia, further studies need to be conducted, not only to improve tumor regression, but also to prevent its collateral damage.
Collapse
Affiliation(s)
- Ana Belén Herrera-Campos
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
| | - Esteban Zamudio-Martinez
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Daniel Delgado-Bellido
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Mónica Fernández-Cortés
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Luis M. Montuenga
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
- Program in Solid Tumors, CIMA-University of Navarra, 31008 Pamplona, Spain
- Navarra Health Research Institute (IDISNA), 31008 Pamplona, Spain
| | - F. Javier Oliver
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| | - Angel Garcia-Diaz
- Instituto de Parasitología y Biomedicina López Neyra, CSIC, 18016 Granada, Spain; (A.B.H.-C.); (E.Z.-M.); (D.D.-B.); (M.F.-C.)
- Consorcio de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain;
| |
Collapse
|
102
|
Pretre V, Papadopoulos D, Regard J, Pelletier M, Woo J. Interleukin-1 (IL-1) and the inflammasome in cancer. Cytokine 2022; 153:155850. [PMID: 35279620 DOI: 10.1016/j.cyto.2022.155850] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/13/2022] [Accepted: 03/03/2022] [Indexed: 12/14/2022]
Abstract
Numerous preclinical and clinical studies have demonstrated the significant contribution of inflammation to the development and progression of various types of cancer. Inflammation in the tumor microenvironment mediates complex interactions between innate immunity, adaptive immunity, microbiomes and stroma, and ultimately alters the overall fitness of tumor cells at multiple stages of carcinogenesis. Malignancies are known to arise in areas of chronic inflammation and inflammation in the tumor microenvironment (often called tumor-promoting inflammation) is believed to allow cancer cells to evade immunosurveillance while promoting genetic instability, survival and progression. Among the strongest data suggesting a causal role for inflammation in cancer come from the recent CANTOS trial which demonstrated that interleukin-1β (IL-1β) inhibition with canakinumab leads to a significant, dose-dependent decrease in incident lung cancer. This observation has launched a series of additional clinical studies to understand the role of IL-1β and the inflammasome in cancer, and the clinical utility of IL-1β inhibition in different stages of lung cancer. In this article we will review recent data implicating IL-1β signaling and its upstream regulator NLRP3 in both solid tumor and hematologic malignancies. We will discuss the key preclinical observations and the current clinical landscape, and describe the pharmacologic tools which will be used to evaluate the effects of blocking tumor-promoting inflammation clinically.
Collapse
|
103
|
Zhou Z, Xu J, Huang N, Tang J, Ma P, Cheng Y. A Pyroptosis-Related Gene Signature Associated with Prognosis and Tumor Immune Microenvironment in Gliomas. Int J Gen Med 2022; 15:4753-4769. [PMID: 35571289 PMCID: PMC9091698 DOI: 10.2147/ijgm.s353762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/16/2022] [Indexed: 11/23/2022] Open
Abstract
Background Pyroptosis is a novel form of cell death that plays a significant role in cancer, while the prognostic values of pyroptosis-related genes in gliomas have not been revealed. Methods We analyzed the RNA-seq and clinical data of gliomas from the University of California Santa Cruz (UCSC) Xena database to determine differentially expressed pyroptosis-related genes. Based on these genes, a pyroptosis genes signature was constructed after univariate Cox analysis and Lasso Cox analyses. The sensitivity and specificity of pyroptosis genes signature were verified by the Chinese Glioma Genome Atlas (CGGA) dataset. Finally, we explored the association of risk signatures with tumor microenvironment and immune cell infiltration. Results Of 15 differentially expressed pyroptosis-related genes, three genes of BCL2 associated X (BAX), caspase 3 (CASP3), and caspase 4 (CASP4) were used to construct the risk signature. The effectiveness of risk signature for predicting survival at 1, 3, 5 years was performed by the receiver operating characteristic curve (ROC), and the area under curves (AUC) was 0.739, 0.817, and 0.800, respectively. Functional enrichment results showed signal transduction, cell adhesion, immune response, and inflammatory response were enriched. The immune analysis revealed that pyroptosis had a remarkable effect on the immune microenvironment. Conclusion In this study, we constructed a pyroptosis-related gene signature, which can serve as a potential biomarker for predicting the survival of glioma patients. Additionally, we suggested that pyroptosis may promote gliomas development by inducing chronic inflammation microenvironment.
Collapse
Affiliation(s)
- Zunjie Zhou
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jing Xu
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ning Huang
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Tang
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ping Ma
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yuan Cheng
- Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Correspondence: Yuan Cheng, Department of Neurosurgery, the Second Affiliated Hospital of Chongqing Medical University, No. 74 Linjiang Road, Yuzhong District, Chongqing, People’s Republic of China, Tel +8613708329653, Email
| |
Collapse
|
104
|
Sun R, Gao DS, Shoush J, Lu B. The IL-1 family in tumorigenesis and antitumor immunity. Semin Cancer Biol 2022; 86:280-295. [DOI: 10.1016/j.semcancer.2022.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
|
105
|
Yuan B, Clowers MJ, Velasco WV, Peng S, Peng Q, Shi Y, Ramos-Castaneda M, Zarghooni M, Yang S, Babcock RL, Chang SH, Heymach JV, Zhang J, Ostrin EJ, Watowich SS, Kadara H, Moghaddam SJ. Targeting IL-1β as an immune preventive and therapeutic modality for K-ras mutant lung cancer. JCI Insight 2022; 7:157788. [PMID: 35471938 PMCID: PMC9220853 DOI: 10.1172/jci.insight.157788] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
K-ras–mutant lung adenocarcinoma (KM-LUAD) is associated with abysmal prognosis and is tightly linked to tumor-promoting inflammation. A human mAb, canakinumab, targeting the proinflammatory cytokine IL-1β, significantly decreased the risk of lung cancer in the Canakinumab Anti-inflammatory Thrombosis Outcomes Study. Interestingly, we found high levels of IL-1β in the lungs of mice with K-rasG12D–mutant tumors (CC-LR mice). Here, we blocked IL-1β using an anti–IL-1β mAb in cohorts of 6- or 14-week-old CC-LR mice to explore its preventive and therapeutic effect, respectively. IL-1β blockade significantly reduced lung tumor burden, which was associated with reprogramming of the lung microenvironment toward an antitumor phenotype characterized by increased infiltration of cytotoxic CD8+ T cells (with high IFN-γ and granzyme B expression but low programmed cell death 1 [PD-1] expression) while suppressing neutrophils and polymorphonuclear (PMN) myeloid-derived suppressor cells. When querying the Cancer Genome Atlas data set, we found positive correlations between IL1B expression and infiltration of immunosuppressive PMNs and expression of their chemoattractant, CXCL1, and PDCD1 expressions in patients with KM-LUAD. Our data provide evidence that IL-1β blockade may be a preventive strategy for high-risk individuals and an alternative therapeutic approach in combination with currently available treatments for KM-LUAD.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Michael J Clowers
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Walter V Velasco
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Stephen Peng
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Qian Peng
- Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Yewen Shi
- Department of Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Marco Ramos-Castaneda
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Melody Zarghooni
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Shuanying Yang
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Rachel L Babcock
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Seon Hee Chang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - John V Heymach
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Jianjun Zhang
- Department of Thoracic Head & Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Edwin J Ostrin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Stephanie S Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Humam Kadara
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| | - Seyed Javad Moghaddam
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, United States of America
| |
Collapse
|
106
|
Lin M, Huang T, Wang X, Li X, Ma J, Su L, Wu J. Non-Canonical NF-κB Signaling Stratifies LGG into Subtypes with Distinct Molecular and Cellular Characteristic and Survival Expectancy. Int J Gen Med 2022; 15:3677-3686. [PMID: 35411180 PMCID: PMC8994666 DOI: 10.2147/ijgm.s347654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/17/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Minhua Lin
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Tianxiang Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
| | - Xuan Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
| | - Xuenan Li
- Beijing Genetron Health, Co. Ltd, Beijing, 102206, People’s Republic of China
| | - Jingjiao Ma
- Beijing Genetron Health, Co. Ltd, Beijing, 102206, People’s Republic of China
| | - Lan Su
- Beijing Genetron Health, Co. Ltd, Beijing, 102206, People’s Republic of China
| | - Jun Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China
- Correspondence: Jun Wu, Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, People’s Republic of China, Tel +86 13508480515, Fax +86 731-89753039, Email
| |
Collapse
|
107
|
Interleukin-38 in Health and Disease. Cytokine 2022; 152:155824. [DOI: 10.1016/j.cyto.2022.155824] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 12/13/2022]
|
108
|
Specific NLRP3 Inflammasome Assembling and Regulation in Neutrophils: Relevance in Inflammatory and Infectious Diseases. Cells 2022; 11:cells11071188. [PMID: 35406754 PMCID: PMC8997905 DOI: 10.3390/cells11071188] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
The NLRP3 inflammasome is a cytosolic multimeric protein platform that leads to the activation of the protease zymogen, caspase-1 (CASP1). Inflammasome activation mediates the proteolytic activation of pro-inflammatory cytokines (IL-1β and IL-18) and program cell death called pyroptosis. The pyroptosis is mediated by the protein executioner Gasdermin D (GSDMD), which forms pores at the plasma membrane to facilitate IL-1β/IL-18 secretion and causes pyroptosis. The NLRP3 inflammasome is activated in response to a large number of pathogenic and sterile insults. However, an uncontrolled inflammasome activation may drive inflammation-associated diseases. Initially, inflammasome-competent cells were believed to be limited to macrophages, dendritic cells (DC), and monocytes. However, emerging evidence indicates that neutrophils can assemble inflammasomes in response to various stimuli with functional relevance. Interestingly, the regulation of inflammasome in neutrophils appears to be unconventional. This review provides a broad overview of the role and regulation of inflammasomes—and more specifically NLRP3—in neutrophils.
Collapse
|
109
|
Xie D, Sun MY. Mechanism of action of NF-κB related cell signaling pathways in progression of gastritis to carcinoma. Shijie Huaren Xiaohua Zazhi 2022; 30:255-259. [DOI: 10.11569/wcjd.v30.i6.255] [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] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is a major global public health problem. The evolvement pattern of "superficial gastritis-chronic atrophic gastritis-intestinal metaplasia-dysplasia-gastric carcinoma" is common in related gastric diseases. As a key factor involved in systemic stress response, inflammatory response, and apoptosis, the regulation of NF-κB related to inflammation and apoptosis is a necessary link between inflammation and cancer progression. NF-κB is activated in most solid tumors and lymphomas. In the critical mechanism of gastric cancer induced by gastritis with various etiologies, the upstream and downstream molecules in the NF-κB signaling pathway are changed, and the cells are exposed to the microenvironment of inflammatory response for a long time, which ultimately leads to the development of their carcinogenic potential. This article discusses the mechanism of NF-κB in the key risk factors for the progression of gastric disease.
Collapse
Affiliation(s)
- Dong Xie
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Chinese Medicine, Shanghai 201203, China
| | - Ming-Yu Sun
- Shuguang Hospital, Key Laboratory of Liver and Kidney Diseases (Ministry of Education), Institute of Liver Diseases, Shanghai University of Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
110
|
Lima de Souza Gonçalves V, Cordeiro Santos ML, Silva Luz M, Santos Marques H, de Brito BB, França da Silva FA, Souza CL, Oliveira MV, de Melo FF. From Helicobacter pylori infection to gastric cancer: Current evidence on the immune response. World J Clin Oncol 2022; 13:186-199. [PMID: 35433296 PMCID: PMC8966509 DOI: 10.5306/wjco.v13.i3.186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 07/31/2021] [Accepted: 02/15/2022] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is the result of a multifactorial process whose main components are infection by Helicobacter pylori (H. pylori), bacterial virulence factors, host immune response and environmental factors. The development of the neoplastic microenvironment also depends on genetic and epigenetic changes in oncogenes and tumor suppressor genes, which results in deregulation of cell signaling pathways and apoptosis process. This review summarizes the main aspects of the pathogenesis of GC and the immune response involved in chronic inflammation generated by H. pylori.
Collapse
Affiliation(s)
| | - Maria Luísa Cordeiro Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Hanna Santos Marques
- Universidade Estadual do Sudoeste da Bahia, Campus Vitória da Conquista, Vitória da Conquista 45083-900, Bahia, Brazil
| | - Breno Bittencourt de Brito
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Cláudio Lima Souza
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Márcio Vasconcelos Oliveira
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
111
|
West AJ, Deswaerte V, West AC, Gearing LJ, Tan P, Jenkins BJ. Inflammasome-Associated Gastric Tumorigenesis Is Independent of the NLRP3 Pattern Recognition Receptor. Front Oncol 2022; 12:830350. [PMID: 35299732 PMCID: PMC8921257 DOI: 10.3389/fonc.2022.830350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/28/2022] [Indexed: 12/24/2022] Open
Abstract
Inflammasomes are important multiprotein regulatory complexes of innate immunity and have recently emerged as playing divergent roles in numerous inflammation-associated cancers. Among these include gastric cancer (GC), the third leading cause of cancer-associated death worldwide, and we have previously discovered a pro-tumorigenic role for the key inflammasome adaptor apoptosis-associated speck-like protein containing a CARD (ASC) in the spontaneous genetic gp130F/F mouse model for GC. However, the identity of the specific pattern recognition receptors (PRRs) that activate tumor-promoting inflammasomes during GC is unknown. Here, we investigated the role of the best-characterized inflammasome-associated PRR, nucleotide-binding domain, and leucine-rich repeat containing receptor, pyrin domain-containing (NLRP) 3, in GC. In gastric tumors of gp130F/F mice, although NLRP3 expression was elevated at the mRNA (qPCR) and protein (immunohistochemistry) levels, genetic ablation of NLRP3 in gp130F/F:Nlrp3-/- mice did not alleviate the development of gastric tumors. Similarly, cellular processes associated with tumorigenesis in the gastric mucosa, namely, proliferation, apoptosis, and inflammation, were comparable between gp130F/F and gp130F/F:Nlrp3-/- mice. Furthermore, inflammasome activation levels, determined by immunoblotting and immunohistochemistry for cleaved Caspase-1, which along with ASC is another integral component of inflammasome complexes, were unchanged in gp130F/F and gp130F/F:Nlrp3-/- gastric tumors. We also observed variable NLRP3 expression levels (mRNA and protein) among independent GC patient cohorts, and NLRP3 was not prognostic for survival outcomes. Taken together, these data suggest that NLRP3 does not play a major role in promoting inflammasome-driven gastric tumorigenesis, and thus pave the way for further investigations to uncover the key inflammasome-associated PRR implicated in GC.
Collapse
Affiliation(s)
- Alice J West
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Virginie Deswaerte
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Alison C West
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Linden J Gearing
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Patrick Tan
- Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore, Singapore.,Genome Institute of Singapore, Singapore, Singapore.,Cancer Sciences Institute of Singapore, National University of Singapore, Institute of Singapore, Singapore, Singapore
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular Translational Science, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
112
|
Barrett's Metaplasia Progression towards Esophageal Adenocarcinoma: An Attempt to Select a Panel of Molecular Sensors and to Reflect Clinical Alterations by Experimental Models. Int J Mol Sci 2022; 23:ijms23063312. [PMID: 35328735 PMCID: PMC8955539 DOI: 10.3390/ijms23063312] [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: 01/03/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
The molecular processes that predispose the development of Barrett’s esophagus (BE) towards esophageal adenocarcinoma (EAC) induced by gastrointestinal reflux disease (GERD) are still under investigation. In this study, based on a scientific literature screening and an analysis of clinical datasets, we selected a panel of 20 genes covering BE- and EAC-specific molecular markers (FZD5, IFNGR1, IL1A, IL1B, IL1R1, IL1RN, KRT4, KRT8, KRT15, KRT18, NFKBIL1, PTGS1, PTGS2, SOCS3, SOX4, SOX9, SOX15, TIMP1, TMEM2, TNFRSF10B). Furthermore, we aimed to reflect these alterations within an experimental and translational in vitro model of BE to EAC progression. We performed a comparison between expression profiles in GSE clinical databases with an in vitro model of GERD involving a BE cell line (BAR-T) and EAC cell lines (OE33 and OE19). Molecular responses of cells treated with acidified bile mixture (BM) at concentration of 100 and 250 μM for 30 min per day were evaluated. We also determined a basal mRNA expression within untreated, wild type cell lines on subsequent stages of BE and EAC development. We observed that an appropriately optimized in vitro model based on the combination of BAR-T, OE33 and OE19 cell lines reflects in 65% and more the clinical molecular alterations observed during BE and EAC development. We also confirmed previous observations that exposure to BM (GERD in vitro) activated carcinogenesis in non-dysplastic cells, inducing molecular alternations in the advanced stages of BE. We conclude that it is possible to induce, to a high extent, the molecular profile observed clinically within appropriately and carefully optimized experimental models, triggering EAC development. This experimental scheme and molecular marker panel might be implemented in further research, e.g., aiming to develop and evaluate novel compounds and prodrugs targeting GERD as well as BE and EAC prevention and treatment.
Collapse
|
113
|
Li Q, Cheng X, Zhou C, Tang Y, Li F, Zhang B, Huang T, Wang J, Tu S. Fruquintinib Enhances the Antitumor Immune Responses of Anti-Programmed Death Receptor-1 in Colorectal Cancer. Front Oncol 2022; 12:841977. [PMID: 35371995 PMCID: PMC8968679 DOI: 10.3389/fonc.2022.841977] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/15/2022] [Indexed: 11/25/2022] Open
Abstract
Background Programmed death receptor-1 (PD-1) blockade shows little benefit in patients with microsatellite-stable colorectal cancer (MSS-CRC). Fruquintinib is a China-made anti-angiogenic drug which is approved for the third line therapy in mCRC. This study investigates the effect of the combination of fruquintinib and PD-1 blockade on MSS-CRC and its relative mechanisms. Methods The mouse allograft tumor models that represent MSS and microsatellite instability (MSI) CRC were established using murine CT26 and MC38 colon cancer cells, respectively, to assess the treatment efficacy. The percentages of immune cells were detected in the peripheral blood, spleen and tumor tissues in the tumor-bearing mice by flow cytometry analysis. Angiogenesis in tumor tissues was detected by immunofluorescence. The safety of drug treatment was evaluated by histopathology analysis in murine main organs. The efficacy of the combination of fruquintinib and sintilimab were verified in the treatment of MSS-CRC patients. Results Our results showed that the combination of fruquintinib and sintilimab exhibited the strongest inhibition of tumor growth and achieved the longest survival time in mice bearing MC38 or CT26 xenograft tumors, compared to fruquintinib and sintilimab alone. Mechanistically, the combination of fruquintinib and sintilimab reduced angiogenesis, reprogramed the vascular structure, enhanced the infiltration of CD8+T cells (p<0.05), CD8+TNFα+ (p<0.05) T cells and CD8+IFNγ+ (p<0.05) T cells and reduced the ratios of MDSCs and macrophages in mice. There was no obvious toxicity observed in the main organs of the tumor-bearing mice with the combined treatment. Moreover, the treatment using the combination of fruquintinib and sintilimab achieved effective response in five patients with refractory advanced MSS CRC. Conclusion Our results show that the combination of fruquintinib and sintilimab greatly inhibits CRC growth by altering tumor immune microenvironment. This study provides the rational for using the combination of fruquintinib and anti-PD-1 antibody for the treatment of advanced CRC.
Collapse
Affiliation(s)
- Qingli Li
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojiao Cheng
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cong Zhou
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yao Tang
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fuli Li
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baiwen Zhang
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tinglei Huang
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianzheng Wang
- Department of Oncology, Henan Cancer Hospital, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Shuiping Tu, ; Jianzheng Wang,
| | - Shuiping Tu
- State Key Laboratory of Oncogenesis and Related Genes, Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Shuiping Tu, ; Jianzheng Wang,
| |
Collapse
|
114
|
Jiang RY, Wang T, Lan QY, Qin YC, Man TT, Sun H, Li ZL, Zhong XT, Mo CM, Rong Z. BuFeiXiaoJiYin ameliorates the NLRP3 inflammation response and gut microbiota in mice with lung cancer companied with Qi-yin deficiency. Cancer Cell Int 2022; 22:121. [PMID: 35292015 PMCID: PMC8922837 DOI: 10.1186/s12935-022-02543-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/07/2022] [Indexed: 01/01/2023] Open
Abstract
Introduction NLRP3 inflammasome responses and gut microbiota have been shown an important role in lung cancer, however, the relationship between gut microbiota and NLRP3 inflammasome responses in lung cancer with Qi-yin deficiency remains elusive. Methods To investigate the effect of the traditional Chinese medicine BuFeiXiaoJiYin (BFXJY) on NLRP3 inflammasome responses and dysbiosis in lung cancer with Qi-yin deficiency, the female BALB/cA-nu mice were treated with LPS and ATP to induce inflammation, and were intragastrically treated with warm Chinese medicine and smoked with shavings to induce Qi-yin deficiency, as well as were injected with 1 × 107/ml A549 cells to simulate lung cancer. Then the three different doses of BuFeiXiaoJiYin (BFXJY) and positive control (CRID3) were used for intervention in mice for 27 consecutive days. Then, we estimated the protection effect of BFXJY on lung cancer mice with Qi-yin deficiency, through deterring tumor growth, NLRP3 inflammasome, PKC signaling, and homeostasis of gut microbiota. Results In this study, we found that BFXJY could inhibit the tumor growth in lung cancer with Qi-yin deficiency by reducing the production of IL-1β and IL-18 and inhibiting NLRP3 inflammasome activation, which might be associated with the inhibition of PKC signaling. Furthermore, BFXJY could promote microbial diversity and balance the microbial composition changes induced by inflammation and Qi-yin deficiency in lung cancer. Conclusion BuFeiXiaoJiYin ameliorates the NLRP3 inflammation response and gut microbiota in mice with lung cancer companied with Qi-yin deficiency. Our study provides a theoretical basis for the clinical development of therapeutic drugs targeting to treat lung cancer.
Collapse
Affiliation(s)
- Rui-Yuan Jiang
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Ting Wang
- Department of Dean's Office, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Qiao-Yu Lan
- Department of Graduate Student, Guangzhou Medical University, No. 1, Xinzao Road, Fanyu District, Guangzhou, 511495, Guangdong, China
| | - Yan-Chun Qin
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Ting-Ting Man
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Hua Sun
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Zhu-Long Li
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Xiao-Ting Zhong
- Department of Graduate Student, Guangxi University of Chinese Medicine, No. 13, Wuhe Road, Qingxiu District, Nanning, 530022, Guangxi, China
| | - Chun-Mei Mo
- Department of Hepatology, Bao'an Authentic TCM Therapy Hospital, No. 99, Laian Road, Baoan District, Shenzhen, 518100, Guangdong, China.
| | - Zhen Rong
- Department of Oncology, Bao'an Authentic TCM Therapy Hospital, No. 99, Laian Road, Baoan District, Shenzhen, 518100, Guangdong, China.
| |
Collapse
|
115
|
Zhang P, Gu Y, Fang H, Cao Y, Wang J, Liu H, Zhang H, Li H, He H, Li R, Lin C, Xu J. Intratumoral IL-1R1 expression delineates a distinctive molecular subset with therapeutic resistance in patients with gastric cancer. J Immunother Cancer 2022; 10:jitc-2021-004047. [PMID: 35110359 PMCID: PMC8811600 DOI: 10.1136/jitc-2021-004047] [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] [Accepted: 01/03/2022] [Indexed: 12/03/2022] Open
Abstract
Background With the essential role of interleukin-1 signaling in cancer-related inflammation, IL-1R1, the main receptor for both IL-1α and IL-1β, demonstrated therapeutic potential in several types of cancer, which has been put into clinical trials. However, the expression profile and critical role of IL-1R1 in gastric cancer (GC) remain obscure. This study aimed to investigate the prognostic significance of IL-1R1 expression and its predictive value for chemotherapy and immunotherapy in GC. Methods The study enrolled three cohorts, consisting of 409 tumor microarray specimens of GC patients from Zhongshan Hospital, 341 transcriptional data from The Cancer Genome Atlas, and 45 transcriptional data from patients treated with pembrolizumab. IL-1R1 mRNA expression was directly acquired from public datasets, and we also detected IL-1R1 protein expression on tumor microarray by immunohistochemistry. Finally, the associations of IL-1R1 expression with clinical outcomes, immune contexture, and genomic features were analyzed. Results High IL-1R1 expression predicted poor prognosis and inferior responsiveness to both 5-fluorouracil-based adjuvant chemotherapy (ACT) and immune checkpoint blockade (ICB). IL-1R1 fostered an immunosuppressive microenvironment characterized by upregulated M2 macrophages and exhausted CD8+ T cells infiltration. Moreover, the expression of IL-1R1 was intrinsically linked to genomic alterations associated with targeted therapies in GC. Conclusions IL-1R1 served as an independent prognosticator and predictive biomarker for ACT and ICB in GC. Furthermore, IL-1R1 antagonists could be a novel agent alone or combined with current therapeutic strategies in GC.
Collapse
Affiliation(s)
- Puran Zhang
- Department of Biochemistry and Molecular Biology, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Yun Gu
- Department of Biochemistry and Molecular Biology, Fudan University School of Basic Medical Sciences, Shanghai, China
| | - Hanji Fang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yifan Cao
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jieti Wang
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Hao Liu
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Heng Zhang
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - He Li
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hongyong He
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Ruochen Li
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Chao Lin
- Department of General Surgery, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jiejie Xu
- Department of Biochemistry and Molecular Biology, Fudan University School of Basic Medical Sciences, Shanghai, China
| |
Collapse
|
116
|
The Immune Underpinnings of Barrett's-Associated Adenocarcinogenesis: a Retrial of Nefarious Immunologic Co-Conspirators. Cell Mol Gastroenterol Hepatol 2022; 13:1297-1315. [PMID: 35123116 PMCID: PMC8933845 DOI: 10.1016/j.jcmgh.2022.01.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 12/10/2022]
Abstract
There is no doubt that chronic gastroesophageal reflux disease increases the risk of esophageal adenocarcinoma (EAC) by several fold (odds ratio, 6.4; 95% CI, 4.6-9.1), and some relationships between reflux disease-mediated inflammation and oncogenic processes have been explored; however, the precise interconnections between the immune response and genomic instabilities underlying these pathologic processes only now are emerging. Furthermore, the precise cell of origin of the precancerous stages associated with EAC development, Barrett's esophagus, be it cardia resident or embryonic remnant, may shape our interpretation of the likely immune drivers. This review integrates the current collective knowledge of the immunology underlying EAC development and outlines a framework connecting proinflammatory pathways, such as those mediated by interleukin 1β, tumor necrosis factor α, leukemia inhibitory factor, interleukin 6, signal transduction and activator of transcription 3, nuclear factor-κB, cyclooxygenase-2, and transforming growth factor β, with oncogenic pathways in the gastroesophageal reflux disease-Barrett's esophagus-EAC cancer sequence. Further defining these immune and molecular railroads may show a map of the routes taken by gastroesophageal cells on their journey toward EAC tumor phylogeny. The selective pressures applied by this immune-induced journey likely impact the phenotype and genotype of the resulting oncogenic destination and further exploration of lesser-defined immune drivers may be useful in future individualized therapies or enhanced selective application of recent immune-driven therapeutics.
Collapse
|
117
|
Nanoparticle-based drug delivery systems in cancer: A focus on inflammatory pathways. Semin Cancer Biol 2022; 86:860-872. [PMID: 35115226 DOI: 10.1016/j.semcancer.2022.01.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 01/23/2022] [Accepted: 01/23/2022] [Indexed: 12/16/2022]
Abstract
It has become necessary to accept the clinical reality of therapeutic agents targeting the cancer-associated immune system. In recent decades, several investigations have highlighted the role of inflammation in cancer development. It has now been recognized that inflammatory cells secrete mediators, including enzymes, chemokines, and cytokines. These secreted substances produce an inflammatory microenvironment that is critically involved in cancer growth. Inflammation may enhance genomic instability leading to DNA damage, activation of oncogenes, or compromised tumor suppressor activity, all of which may promote various phases of carcinogenesis. Conventional cancer treatment includes surgery, radiation, and chemotherapy. However, treatment failure occurs because current strategies are unable to achieve complete local control due to metastasis. Nanoparticles (NPs) are a broad spectrum of drug carriers typically below the size of 100 nm, targeting tumor sites while reducing off-target consequences. More importantly, NPs can stimulate innate and adaptive immune systems in the tumor microenvironment (TME); hence, they induce a cancer-fighting immune response. Strikingly, targeting cancer cells with NPs helps eliminate drug resistance and tumor recurrence, as well as prevents inflammation. Throughout this review, we provide recent data on the role of inflammation in cancer and explore nano-therapeutic initiatives to target significant mediators, for example, nuclear factor-kappa B (NF-κB), tumor necrosis factor-α (TNF-α), and interleukins (ILs) associated with cancer-related inflammation, to escort the immunomodulators to cancer cells and associated systemic compartments. We also highlight the necessity of better identifying inflammatory pathways in cancer pathophysiology to develop effective treatment plans.
Collapse
|
118
|
Wang L, Hu D, Xie B, Xie L. Blockade of Myd88 signaling by a novel MyD88 inhibitor prevents colitis-associated colorectal cancer development by impairing myeloid-derived suppressor cells. Invest New Drugs 2022; 40:506-518. [PMID: 35089465 PMCID: PMC9098617 DOI: 10.1007/s10637-022-01218-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/21/2022] [Indexed: 01/04/2023]
Abstract
Background. In cancer, myeloid-derived suppressor cells (MDSCs) are known to escape the host immune system by developing a highly suppressive environment. However, little is known about the molecular mechanism behind MDSC-mediated tumor cell evasion of the immune system. Toll-like receptor (TLR) signaling elicited in the tumor microenvironment has the potential to induce MDSC differentiations in different organs. Therefore, MDSC elimination by blocking the action of myeloid differentiation factor 88 (MyD88), which is a key adaptor-signaling molecule that affects TLR activity, seems to be an ideal tumor immunotherapy. Previous studies have proven that blocking MyD88 signaling with a novel MyD88 inhibitor (TJ-M2010-5, synthesized by Zhou’s group) completely prevented colitis-associated colorectal cancer (CAC) development in mice. Methods. In the present study, we investigated the impact of the novel MyD88 inhibitor on the number, phenotype, and function of MDSC in the mice model of CAC. Results. We showed that CAC growth inhibition was involved in diminished MDSC generation, expansion, and suppressive function and that MDSC-mediated immune escape was dependent on MyD88 signaling pathway activation. MyD88 inhibitor treatment decreased the accumulation of CD11b+Gr1+ MDSCs in mice with CAC, thereby reducing cytokine (GM-CSF, G-CSF, IL-1β, IL-6 and TGF-β) secretion associated with MDSC accumulation, and reducing the expression of molecules (iNOS, Arg-1 and IDO) associated with the suppressive capacity of MDSCs. In addition, MyD88 inhibitor treatment reduced the differentiation of MDSCs from myeloid cells and the suppressive capacity of MDSCs on the proliferation of activated CD4+ T cells in vitro. Conclusion. MDSCs are primary cellular targets of a novel MyD88 inhibitor during CAC development. Our findings prove that MyD88 signaling is involved in the regulation of the immunosuppressive functions of MDSCs. The novel MyD88 inhibitor TJ-M2010-5 is a new and effective agent that modulates MyD88 signaling to overcome MDSC suppressive functions, enabling the development of successful antitumor immunotherapy.
Collapse
Affiliation(s)
- Lu Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Dan Hu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bin Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Lin Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
119
|
Lin J, Lai X, Liu X, Yan H, Wu C. Pyroptosis in glioblastoma: A crucial regulator of the tumour immune microenvironment and a predictor of prognosis. J Cell Mol Med 2022; 26:1579-1593. [PMID: 35083859 PMCID: PMC8899201 DOI: 10.1111/jcmm.17200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/30/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Recent studies have shown that pyroptosis, an inflammatory form of cell death, has a dual role in tumorigenesis and tumour progression and affects the prognosis of patients; however, the role of pyroptosis in glioblastoma (GBM) is still unclear. In this study, based on GBM patients' data from two independent cohorts, we performed a comprehensive analysis of the expression and prognostic value of 33 pyroptosis‐associated genes (PAGs) in GBM, as well as their role in the tumour immune microenvironment (TIME) of GBM. We identified 29 PAGs that were differentially expressed between GBM and normal brain tissue, 18 of which were upregulated in GBM tissue. Most of the 33 PAGs were strongly correlated with the levels of immune cell infiltration. Based on the 33 PAGs, the GBM samples can be divided into two clusters (C1‐C2), with C1 having a ‘hot’ but immunosuppressive TIME and C2 having a ‘cold’ TIME, suggesting different immunotherapeutic responses in the two clusters. In addition, we identified four PAGs that were strongly associated with GBM prognosis and constructed a risk model based on these four PAGs. This risk model is an independent prognostic factor for GBM patients, and there is a different immune status between high‐ and low‐risk groups. In conclusion, this study demonstrates that pyroptosis is closely associated with the prognosis and TIME of GBM and provides an important basis for further studies on the relationship between pyroptosis and GBM.
Collapse
Affiliation(s)
- Jinhu Lin
- Department of Neurosurgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Xiang Lai
- Department of Neurosurgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Xiaoping Liu
- Department of Neurosurgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Hua Yan
- Department of Neurosurgery, Meizhou People's Hospital (Huangtang Hospital), Meizhou, China
| | - Changwu Wu
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
120
|
Lokman MS, Zaafar D, Althagafi HA, Abdel Daim MM, Theyab A, Hasan Mufti A, Algahtani M, Habotta OA, Alghamdi AAA, Alsharif KF, Albrakati A, Oyouni AAA, Bauomy AA, Baty RS, Zhery AS, Hassan KE, Abdel Moneim AE, Kassab RB. Antiulcer activity of proanthocyanidins is mediated via suppression of oxidative, inflammatory, and apoptotic machineries. J Food Biochem 2022; 46:e14070. [PMID: 35034361 DOI: 10.1111/jfbc.14070] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/24/2022]
Abstract
Gastric ulcer (GU) is a lesion in the gastric mucosa associated with excessive oxidative damage, inflammatory response, apoptotic events, and irritation which may develop into cancer. However, medications commonly used in GU treatment cannot normalize gastric mucosa, while causing several adverse effects. Proanthocyanidins (PAs) are dietary flavonoids with numerous biological and pharmacological activities. In the current investigation, we studied the potential anti-ulcerative activity of PAs against acidified ethanol (HCl/ethanol)-caused gastric ulceration. Fifty male albino Wistar rats were allocated into five equal groups: control, HCl/ethanol (3 mL/kg), lansoprazole (LPZ, 30 mg/kg) + HCl/ethanol, and PAs (100 and 250 mg/kg) + HCl/ethanol. LPZ and PAs were applied one week before gastric ulcer induction. PAs pretreatment notably reduced gastric mucosal macroscopic and microscopic pathological changes in a dose-dependent manner. Additionally, PAs activated the innate antioxidant molecules including glutathione and its derived antioxidants (glutathione peroxidase and glutathione reductase), along with superoxide dismutase and catalase, while attenuating pro-oxidant formation, including malondialdehyde and nitric oxide. Interestingly, PAs supplementation at a higher dose suppressed gastric inflammatory and apoptotic responses, as demonstrated by the reduced levels of interleukin-1β, interleukin-6, tumor necrosis factor alpha, high-mobility group box 1, cyclooxygenase 2, prostaglandin E2, nuclear factor kappa-B, Bcl-2-associated X protein, and caspase-3, while B cell lymphoma 2 was elevated. Hence, PAs could exhibit antiulcer activity by protecting gastric tissue from the development of oxidative damage, inflammatory responses, and apoptosis events associated with ulceration. PRACTICAL IMPLICATIONS: Gastric ulcer is a lesion in the gastric mucosal layer associated with excessive inflammatory response, apoptotic events, oxidative damage, and irritation, and may develop into cancer with about 5%-10% morbidity rate. However, medications commonly used in GU treatment cannot normalize gastric mucosa, while causing several adverse effects. Therefore, new therapeutic approaches are needed to treat or prevent gastric ulceration. Proanthocyanidins (PAs, condensed tannins) are dietary flavonoids found in abundance in different plant species, including their fruits, bark, and seeds. Due to their potent antioxidative activity, PAs have been applied to prevent or treat oxidative stress-related diseases, including cancer, as well as metabolic, neurodegenerative, cardiovascular, and inflammatory disorders. Here, we examine the potential therapeutic role of proanthocyanidins (PAs) against acidified ethanol-induced gastric ulcer in rats through evaluating oxidative challenge, inflammatory response, apoptotic events, and histopathological changes in the gastric tissue.
Collapse
Affiliation(s)
- Maha S Lokman
- Biology Department, College of Science and Humanities, Prince Sattam bin Abdul Aziz University, Alkharj, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Dalia Zaafar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Hussam A Althagafi
- Department of Biology, Faculty of Science and Arts, Al-Baha University, Almakhwah, Saudi Arabia
| | - Mohamed M Abdel Daim
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Abdulrahman Theyab
- Department of Laboratory Medicine, Security Forces Hospital, Mecca, Saudi Arabia
| | - Ahmad Hasan Mufti
- Medical Genetics Department, Faculty of Medicine, Umm Al-Qura University, Saudi Arabia
| | - Mohammad Algahtani
- Department of Clinical Laboratories Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ola A Habotta
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | | | - Khalaf F Alsharif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ashraf Albrakati
- Department of Human Anatomy, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Atif Abdulwahab A Oyouni
- Department of Biology, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia.,Genome and Biotechnology Unit, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Amira A Bauomy
- Department of Science Laboratories, College of Science and Arts, Qassim University, Saudi Arabia
| | - Roua S Baty
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Ahmed S Zhery
- Kasr Al-Eini School of Medicine, Cairo University, Cairo, Egypt
| | - Khalid E Hassan
- Department of Pathology, College of Medicine, Taif University, Taif, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt.,Department of Biology, Faculty of Science and Arts, Al-Baha University, Almakhwah, Saudi Arabia
| |
Collapse
|
121
|
Fu X, De Angelis C, Schiff R. Interferon Signaling in Estrogen Receptor-positive Breast Cancer: A Revitalized Topic. Endocrinology 2022; 163:6429717. [PMID: 34791151 DOI: 10.1210/endocr/bqab235] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Indexed: 12/25/2022]
Abstract
Cancer immunology is the most rapidly expanding field in cancer research, with the importance of immunity in cancer pathogenesis now well accepted including in the endocrine-related cancers. The immune system plays an essential role in the development of ductal and luminal epithelial differentiation in the mammary gland. Originally identified as evolutionarily conserved antipathogen cytokines, interferons (IFNs) have shown important immune-modulatory and antineoplastic properties when administered to patients with various types of cancer, including breast cancer. Recent studies have drawn attention to the role of tumor- and stromal-infiltrating lymphocytes in dictating therapy response and outcome of breast cancer patients, which, however, is highly dependent on the breast cancer subtype. The emerging role of tumor cell-inherent IFN signaling in the subtype-defined tumor microenvironment could influence therapy response with protumor activities in breast cancer. Here we review evidence with new insights into tumor cell-intrinsic and tumor microenvironment-derived IFN signaling, and the crosstalk of IFN signaling with key signaling pathways in estrogen receptor-positive (ER+) breast cancer. We also discuss clinical implications and opportunities exploiting IFN signaling to treat advanced ER+ breast cancer.
Collapse
Affiliation(s)
- Xiaoyong Fu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Carmine De Angelis
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80138 Naples, Italy
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
122
|
Vantucci CE, Guyer T, Leguineche K, Chatterjee P, Lin A, Nash KE, Hastings MA, Fulton T, Smith CT, Maniar D, Frey Rubio DA, Peterson K, Harrer JA, Willett NJ, Roy K, Guldberg RE. Systemic Immune Modulation Alters Local Bone Regeneration in a Delayed Treatment Composite Model of Non-Union Extremity Trauma. Front Surg 2022; 9:934773. [PMID: 35874126 PMCID: PMC9300902 DOI: 10.3389/fsurg.2022.934773] [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: 05/03/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022] Open
Abstract
Bone non-unions resulting from severe traumatic injuries pose significant clinical challenges, and the biological factors that drive progression towards and healing from these injuries are still not well understood. Recently, a dysregulated systemic immune response following musculoskeletal trauma has been identified as a contributing factor for poor outcomes and complications such as infections. In particular, myeloid-derived suppressor cells (MDSCs), immunosuppressive myeloid-lineage cells that expand in response to traumatic injury, have been highlighted as a potential therapeutic target to restore systemic immune homeostasis and ultimately improve functional bone regeneration. Previously, we have developed a novel immunomodulatory therapeutic strategy to deplete MDSCs using Janus gold nanoparticles that mimic the structure and function of antibodies. Here, in a preclinical delayed treatment composite injury model of bone and muscle trauma, we investigate the effects of these nanoparticles on circulating MDSCs, systemic immune profiles, and functional bone regeneration. Unexpectedly, treatment with the nanoparticles resulted in depletion of the high side scatter subset of MDSCs and an increase in the low side scatter subset of MDSCs, resulting in an overall increase in total MDSCs. This overall increase correlated with a decrease in bone volume (P = 0.057) at 6 weeks post-treatment and a significant decrease in mechanical strength at 12 weeks post-treatment compared to untreated rats. Furthermore, MDSCs correlated negatively with endpoint bone healing at multiple timepoints. Single cell RNA sequencing of circulating immune cells revealed differing gene expression of the SNAb target molecule S100A8/A9 in MDSC sub-populations, highlighting a potential need for more targeted approaches to MDSC immunomodulatory treatment following trauma. These results provide further insights on the role of systemic immune dysregulation for severe trauma outcomes in the case of non-unions and composite injuries and suggest the need for additional studies on targeted immunomodulatory interventions to enhance healing.
Collapse
Affiliation(s)
- Casey E Vantucci
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Tyler Guyer
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Kelly Leguineche
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Paramita Chatterjee
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America.,Marcus Center for Therapeutic Cell Characterization and Manufacturing, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Angela Lin
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Kylie E Nash
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Molly Ann Hastings
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Travis Fulton
- The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, United States of America.,Department of Orthopaedics, Emory University, Atlanta, GA, United States of America
| | - Clinton T Smith
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Drishti Maniar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - David A Frey Rubio
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Kaya Peterson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America
| | - Julia Andraca Harrer
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| | - Nick J Willett
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America.,The Atlanta Veterans Affairs Medical Center Atlanta, Decatur, GA, United States of America.,Department of Orthopaedics, Emory University, Atlanta, GA, United States of America
| | - Krishnendu Roy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States of America.,Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, United States of America
| | - Robert E Guldberg
- Knight Campus or Accelerating Scientific Impact, University of Oregon, Eugene, OR, United States of America
| |
Collapse
|
123
|
Abstract
The intimate involvement of pathogens with the heightened risk for developing certain cancers is an area of research that has captured a great deal of attention over the last 10 years. One firmly established paradigm that highlights this aspect of disease progression is in the instance of Helicobacter pylori infection and the contribution it makes in elevating the risk for developing gastric cancer. Whilst the molecular mechanisms that pinpoint the contribution that this microorganism inflicts towards host cells during gastric cancer initiation have come into greater focus, another picture that has also emerged is one that implicates the host's immune system, and the chronic inflammation that can arise therefrom, as being a central contributory factor in disease progression. Consequently, when taken with the underlying role that the extracellular matrix plays in the development of most cancers, and how this dynamic can be modulated by proteases expressed from the tumor or inflammatory cells, a complex and detailed relationship shared between the individual cellular components and their surroundings is coming into focus. In this review article, we draw attention to the emerging role played by the cathepsin proteases in modulating the stage-specific progression of Helicobacter pylori-initiated gastric cancer and the underlying immune response, while highlighting the therapeutic significance of this dynamic and how it may be amenable for novel intervention strategies within a basic research or clinical setting.
Collapse
|
124
|
Study of the correlation between the expression of nuclear factor kappa B and proliferation regulatory proteins and chronic superficial gastritis. VOJNOSANIT PREGL 2022. [DOI: 10.2298/vsp200807135h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Background/Aim. Cell proliferation and the regulation of protein expression play an important role in gastritis, but in chronic superficial gastritis (CSG), they are rarely reported. The aim of this study was to determine the relationship between the expression of nuclear factor kappa B (NF-?B) and regulatory proteins and the rat CSG. Methods. The CSG rat model was established artificially, by chemical agents and irregular diet. The expression of epidermal growth factor receptor (EGFR) and proliferating cell nuclear antigen (PCNA) in the gastric mucosa of CSG rats was measured by immunohistochemistry, while mRNA expression levels of NF-?B p65 were detected by in situ hybridization. Results. There was more obvious infiltration of inflammatory cells in the gastric mucosa of CSG rats than in that of control rats, and the inflammation score was significantly increased. The expression levels of PCNA, EGFR, and NF-?B p65 mRNA in the gastric mucosal cells of CSG model rats increased significantly. Correlation analysis showed that the inflammation score was positively correlated with the expression levels of NF-?B p65 mRNA and EGFR, while it presented no significant correlation with the expression level of PCNA. In addition, there was a significant positive correlation between NF-?B p65 mRNA and EGFR levels. Conclusion. High expression of NF-?B and EGFR plays an important role in the occurrence and progression of CSG, and it is significantly positively correlated with the degree of inflammation in the gastric mucosa. Therefore, changes in NF-?B and EGFR expression may be used as important indicators for the assessment of CSG; changes in their expression levels are helpful to assess the degree of gastric mucosal lesions and progression of CSG.
Collapse
|
125
|
Dosch AR, Singh S, Nagathihalli NS, Datta J, Merchant NB. Interleukin-1 signaling in solid organ malignancies. Biochim Biophys Acta Rev Cancer 2021; 1877:188670. [PMID: 34923027 DOI: 10.1016/j.bbcan.2021.188670] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/19/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
As inflammation plays a critical role in the development and progression of cancer, therapeutic targeting of cytokine pathways involved in both tumorigenesis and dictating response to clinical treatments are of significant interest. Recent evidence has highlighted the importance of the pro-inflammatory cytokine interleukin-1 (IL-1) as a key mediator of tumor growth, metastatic disease spread, immunosuppression, and drug resistance in cancer. IL-1 promotes tumorigenesis through diverse mechanisms, including the activation of oncogenic signaling pathways directly in tumor cells and via orchestrating crosstalk between the cellular constituents of the tumor microenvironment (TME), thereby driving cancer growth. This review will provide an overview of IL-1 signaling and physiology and summarize the disparate mechanisms involving IL-1 in tumorigenesis and cancer progression. Additionally, clinical studies targeting IL-1 signaling in the management of solid organ tumors will be summarized herein.
Collapse
Affiliation(s)
- Austin R Dosch
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Samara Singh
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nagaraj S Nagathihalli
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Jashodeep Datta
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America
| | - Nipun B Merchant
- Division of Surgical Oncology, Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, United States of America; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, United States of America.
| |
Collapse
|
126
|
Shi H, Qin Y, Tian Y, Wang J, Wang Y, Wang Z, Lv J. Interleukin-1beta triggers the expansion of circulating granulocytic myeloid-derived suppressor cell subset dependent on Erk1/2 activation. Immunobiology 2021; 227:152165. [PMID: 34936966 DOI: 10.1016/j.imbio.2021.152165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/08/2021] [Accepted: 12/11/2021] [Indexed: 11/18/2022]
Abstract
Chronic inflammation contributes to cancer development and progression. Although interleukin-1beta (IL-1β) has been observed to be associated with an general immune suppression of T cell response and the immunosuppression strongly correlates with accumulation of myeloid-derived suppressor cells (MDSCs), the relationship and mechanism between MDSCs expansion and IL-1β expression remain ambiguous. Here, we showed that the concentration of IL-1β was highly correlated with G-MDSC subset, rather than mo-MDSC subset. Recombinant IL-1β increased the percentage of G-MDSCs in the blood of tumor-bearing mice, and IL-1Ra attenuated the accumulation of G-MDSCs in the tumor-bearing mice. In addition, the IL-1β-overexpressing B16F10 cells induced higher level of G-MDSCs compared with wild-type B16F10 cells. Moreover, we found that the accumulation of G-MDSCs induced by IL-1β was dependent on the activation of extracellular signal-regulated kinases 1 and 2 (Erk1/2). Collectively, these findings show a novel role of IL-1β in G-MDSCs accumulation by activating Erk1/2, which suggests that IL-1β elimination or Erk1/2 signaling blockade could decrease G-MDSCs generation and thereby improve host immunosurveillance.
Collapse
Affiliation(s)
- Huifang Shi
- Clinical Laboratory, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China.
| | - Yan Qin
- Clinical Laboratory, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China
| | - Yufeng Tian
- Clinical Laboratory, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China
| | - Jiaan Wang
- Department of Blood Transfusion, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China
| | - Yan Wang
- Department of Medical Image, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China
| | - Ziyi Wang
- Department of Anesthesiology, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China
| | - Jie Lv
- Clinical Laboratory, The Rizhao People's Hospital Affiliated to Jining Medical University, Rizhao, Shandong, China.
| |
Collapse
|
127
|
Shan C, Li M, Liu Z, Xu R, Qiao F, Du ZY, Zhang ML. Pediococcus pentosaceus Enhances Host Resistance Against Pathogen by Increasing IL-1β Production: Understanding Probiotic Effectiveness and Administration Duration. Front Immunol 2021; 12:766401. [PMID: 34899717 PMCID: PMC8662542 DOI: 10.3389/fimmu.2021.766401] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
Probiotic administration is a potential strategy against enteric pathogen infection in either clinical treatment or animal nutrition industry, but the administration duration of probiotics varied and the underlying mechanisms remain unclear. A strain (YC) affiliated to Pediococcus pentosaceus, a commonly used probiotic, was isolated from fish gut and the potential role of YC against Aeromonas hydrophila was detected in zebrafish. We found that 3- or 4-week YC administration (YC3W or YC4W) increased the resistance against A. hydrophila while 1- or 2-week treatment (YC1W or YC2W) did not. To determine the possible reason, intestinal microbiota analysis and RNAseq were conducted. The results showed that compared with CON and YC1W, YC4W significantly increased the abundance of short-chain fatty acids (SCFAs) producing bacteria and elevated the gene expression of nlrp3. Higher butyrate content and enhanced expression of IL1β were subsequently found in YC4W. To identify the causal relationship between butyrate and the higher pathogen resistance, different concentrations of sodium butyrate (SB) were supplemented. The results suggested that 10 mmol/kg SB addition mirrored the protective effect of YC4W by increasing the production of IL-1β. Furthermore, the increased IL-1β raised the percentage of intestinal neutrophils, which endued the zebrafish with A. hydrophila resistance. In vivo knockdown of intestinal il1b eliminated the anti-infection effect. Collectively, our data suggested that the molecular mechanism of probiotics determined the administration duration, which is vital for the efficiency of probiotics. Promoting host inflammation by probiotic pretreatment is one potential way for probiotics to provide their protective effects against pathogens.
Collapse
Affiliation(s)
- Chengjie Shan
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Miao Li
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhu Liu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China.,School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Rong Xu
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health, School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|
128
|
Tan Q, Duan L, Huang Q, Chen W, Yang Z, Chen J, Jin Y. Interleukin -1β Promotes Lung Adenocarcinoma Growth and Invasion Through Promoting Glycolysis via p38 Pathway. J Inflamm Res 2021; 14:6491-6509. [PMID: 34880649 PMCID: PMC8648110 DOI: 10.2147/jir.s319433] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background There is a close relationship among inflammation, glycolysis, and tumors. The IL-1 family includes important inflammatory cytokines, among which IL-1β has been widely studied. In this study, we focused on the effect of IL-1β on glycolysis of lung adenocarcinoma (LUAD) cells in vivo and in vitro and explored its possible mechanisms. Methods A bioinformatic database and quantitative real-time PCR were used to analyze the expression of glycolysis-related enzyme genes and their correlations with IL1β in human LUAD samples. The human LUAD cell line A549 and Lewis lung carcinoma LLC cell line were stimulated with IL-1β. In vitro treatment effects, including glycolysis level, migration, and invasion were evaluated with a glucose assay kit, lactate assay kit, Western blotting, wound healing, and the transwell method. We established a mouse model of subcutaneous tumors using LLC cells pretreated with IL-1β and analyzed in vivo treatment effects through positron-emission tomography-computed tomography and staining. Virtual screening and molecular dynamic simulation were used to screen potential inhibitors of IL-1β. Results Our results showed that IL1β was positively correlated with the expression of glycolysis-related enzyme genes in LUAD. Glycolysis, migration, and invasion significantly increased in A549 and LLC stimulated with IL-1β. In vivo, IL-1β increased growth, mean standard uptake value, and pulmonary tumor metastasis, which were inhibited by the glycolysis inhibitor 2-deoxy-D-glucose and p38-pathway inhibitors. Small molecular compound ZINC14610053 was suggested being a potential inhibitor of IL-1β. Conclusion IL-1β promotes glycolysis of LUAD cells through p38 signaling, further enhancing tumor-cell migration and invasion. These results show that IL-1β links inflammation to glycolysis in LUAD, and targeting IL-1β and the glycolysis pathway may be a potential therapeutic strategy for lung cancer.
Collapse
Affiliation(s)
- Qi Tan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Limin Duan
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Qi Huang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Wenjuan Chen
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Zimo Yang
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Jiangbin Chen
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases of Health Ministry, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, Hubei, People's Republic of China
| |
Collapse
|
129
|
Leiva O, AbdelHameid D, Connors JM, Cannon CP, Bhatt DL. Common Pathophysiology in Cancer, Atrial Fibrillation, Atherosclerosis, and Thrombosis: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2021; 3:619-634. [PMID: 34988471 PMCID: PMC8702799 DOI: 10.1016/j.jaccao.2021.08.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease and cancer are the 2 leading causes of death worldwide. Emerging evidence suggests common mechanisms between cancer and cardiovascular disease, including atrial fibrillation and atherosclerosis. With advances in cancer therapies, screening, and diagnostics, cancer-specific survival and outcomes have improved. This increase in survival has led to the coincidence of cardiovascular disease, including atrial fibrillation and atherosclerosis, as patients with cancer live longer. Additionally, cancer and cardiovascular disease share several risk factors and underlying pathophysiologic mechanisms, including inflammation, cancer-related factors including treatment effects, and alterations in platelet function. Patients with cancer are at increased risk for bleeding and thrombosis compared with the general population. Although optimal antithrombotic therapy, including agent choice and duration, has been extensively studied in the general population, this area remains understudied in patients with cancer despite their altered thrombotic and bleeding risk. Future investigation, including incorporation of cancer-specific characteristics to traditional thrombotic and bleeding risk scores, clinical trials in the cancer population, and the development of novel antithrombotic and anti-inflammatory strategies on the basis of shared pathophysiologic mechanisms, is warranted to improve outcomes in this patient population.
Collapse
Key Words
- AF, atrial fibrillation
- CAD, coronary artery disease
- CHIP, clonal hematopoiesis of indeterminate potential
- CI, confidence interval
- CLEC-2, C-type lectin-like receptor 2
- HR, hazard ratio
- IL, interleukin
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- ROS, reactive oxygen species
- TKI, tyrosine kinase inhibitor
- VTE, venous thromboembolism
- arrhythmia
- risk factor
- thrombosis
Collapse
Affiliation(s)
- Orly Leiva
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Duaa AbdelHameid
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Jean M. Connors
- Division of Hematology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher P. Cannon
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Deepak L. Bhatt
- Brigham and Women’s Hospital Heart & Vascular Center and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
130
|
Zhang T, Ma C, Zhang Z, Zhang H, Hu H. NF-κB signaling in inflammation and cancer. MedComm (Beijing) 2021; 2:618-653. [PMID: 34977871 PMCID: PMC8706767 DOI: 10.1002/mco2.104] [Citation(s) in RCA: 181] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 02/06/2023] Open
Abstract
Since nuclear factor of κ-light chain of enhancer-activated B cells (NF-κB) was discovered in 1986, extraordinary efforts have been made to understand the function and regulating mechanism of NF-κB for 35 years, which lead to significant progress. Meanwhile, the molecular mechanisms regulating NF-κB activation have also been illuminated, the cascades of signaling events leading to NF-κB activity and key components of the NF-κB pathway are also identified. It has been suggested NF-κB plays an important role in human diseases, especially inflammation-related diseases. These studies make the NF-κB an attractive target for disease treatment. This review aims to summarize the knowledge of the family members of NF-κB, as well as the basic mechanisms of NF-κB signaling pathway activation. We will also review the effects of dysregulated NF-κB on inflammation, tumorigenesis, and tumor microenvironment. The progression of the translational study and drug development targeting NF-κB for inflammatory diseases and cancer treatment and the potential obstacles will be discussed. Further investigations on the precise functions of NF-κB in the physiological and pathological settings and underlying mechanisms are in the urgent need to develop drugs targeting NF-κB for inflammatory diseases and cancer treatment, with minimal side effects.
Collapse
Affiliation(s)
- Tao Zhang
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Chao Ma
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Zhiqiang Zhang
- Immunobiology and Transplant Science CenterHouston Methodist HospitalHoustonTexasUSA
| | - Huiyuan Zhang
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| | - Hongbo Hu
- Cancer Center and Center for Immunology and HematologyWest China HospitalSichuan UniversityChengduSichuanChina
| |
Collapse
|
131
|
Duan S, Rico K, Merchant JL. Gastrin: From Physiology to Gastrointestinal Malignancies. FUNCTION (OXFORD, ENGLAND) 2021; 3:zqab062. [PMID: 35330921 PMCID: PMC8788842 DOI: 10.1093/function/zqab062] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/14/2021] [Accepted: 11/16/2021] [Indexed: 01/07/2023]
Abstract
Abetted by widespread usage of acid-suppressing proton pump inhibitors (PPIs), the mitogenic actions of the peptide hormone gastrin are being revisited as a recurring theme in various gastrointestinal (GI) malignancies. While pathological gastrin levels are intricately linked to hyperplasia of enterochromaffin-like cells leading to carcinoid development, the signaling effects exerted by gastrin on distinct cell types of the gastric mucosa are more nuanced. Indeed, mounting evidence suggests dichotomous roles for gastrin in both promoting and suppressing tumorigenesis. Here, we review the major upstream mediators of gastrin gene regulation, including inflammation secondary to Helicobacter pylori infection and the use of PPIs. We further explore the molecular biology of gastrin in GI malignancies, with particular emphasis on the regulation of gastrin in neuroendocrine neoplasms. Finally, we highlight tissue-specific transcriptional targets as an avenue for targetable therapeutics.
Collapse
Affiliation(s)
- Suzann Duan
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Karen Rico
- Department of Medicine, Division of Gastroenterology and Hepatology, Arizona Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | | |
Collapse
|
132
|
Hou J, Hsu JM, Hung MC. Molecular mechanisms and functions of pyroptosis in inflammation and antitumor immunity. Mol Cell 2021; 81:4579-4590. [PMID: 34562371 DOI: 10.1016/j.molcel.2021.09.003] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/17/2021] [Accepted: 08/31/2021] [Indexed: 12/12/2022]
Abstract
Canonically, gasdermin D (GSDMD) cleavage by caspase-1 through inflammasome signaling triggers immune cell pyroptosis (ICP) as a host defense against pathogen infection. However, cancer cell pyroptosis (CCP) was recently discovered to be activated by distinct molecular mechanisms in which GSDMB, GSDMC, and GSDME, rather than GSDMD, are the executioners. Moreover, instead of inflammatory caspases, apoptotic caspases and granzymes are required for gasdermin protein cleavage to induce CCP. Sufficient accumulation of protease-cleaved gasdermin proteins is the prerequisite for CCP. Inflammation induced by ICP or CCP results in diametrically opposite effects on antitumor immunity because of the differential duration and released cellular contents, leading to contrary effects on therapeutic outcomes. Here, we focus on the distinct mechanisms of ICP and CCP and discuss the roles of ICP and CCP in inflammation and antitumor immunity, representing actionable targets.
Collapse
Affiliation(s)
- Junwei Hou
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, Hunan, China; Otolaryngology Major Disease Research Key Laboratory of Hunan Province, Xiangya Road 87, Changsha 410008, Hunan, China; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Xiangya Road 87, Changsha 410008, Hunan, China; Department of Molecular and Cellular Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jung-Mao Hsu
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung, Taiwan; Department of Molecular and Cellular Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
133
|
Van Den Eeckhout B, Huyghe L, Van Lint S, Burg E, Plaisance S, Peelman F, Cauwels A, Uzé G, Kley N, Gerlo S, Tavernier J. Selective IL-1 activity on CD8 + T cells empowers antitumor immunity and synergizes with neovasculature-targeted TNF for full tumor eradication. J Immunother Cancer 2021; 9:jitc-2021-003293. [PMID: 34772757 PMCID: PMC8593706 DOI: 10.1136/jitc-2021-003293] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 01/31/2023] Open
Abstract
Background Clinical success of therapeutic cancer vaccines depends on the ability to mount strong and durable antitumor T cell responses. To achieve this, potent cellular adjuvants are highly needed. Interleukin-1β (IL-1β) acts on CD8+ T cells and promotes their expansion and effector differentiation, but toxicity and undesired tumor-promoting side effects hamper efficient clinical application of this cytokine. Methods This ‘cytokine problem’ can be solved by use of AcTakines (Activity-on-Target cytokines), which represent fusions between low-activity cytokine mutants and cell type-specific single-domain antibodies. AcTakines deliver cytokine activity to a priori selected cell types and as such evade toxicity and unwanted off-target side effects. Here, we employ subcutaneous melanoma and lung carcinoma models to evaluate the antitumor effects of AcTakines. Results In this work, we use an IL-1β-based AcTakine to drive proliferation and effector functionality of antitumor CD8+ T cells without inducing measurable toxicity. AcTakine treatment enhances diversity of the T cell receptor repertoire and empowers adoptive T cell transfer. Combination treatment with a neovasculature-targeted tumor necrosis factor (TNF) AcTakine mediates full tumor eradication and establishes immunological memory that protects against secondary tumor challenge. Interferon-γ was found to empower this AcTakine synergy by sensitizing the tumor microenvironment to TNF. Conclusions Our data illustrate that anticancer cellular immunity can be safely promoted with an IL-1β-based AcTakine, which synergizes with other immunotherapies for efficient tumor destruction.
Collapse
Affiliation(s)
- Bram Van Den Eeckhout
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Leander Huyghe
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Sandra Van Lint
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Elianne Burg
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | - Frank Peelman
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Anje Cauwels
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Gilles Uzé
- IRMB, University Montpellier, INSERM, CNRS, Montpellier, France
| | - Niko Kley
- Orionis Biosciences Inc, Waltham, Massachusetts, USA
| | - Sarah Gerlo
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium .,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium .,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium.,Orionis Biosciences Inc, Waltham, Massachusetts, USA
| |
Collapse
|
134
|
Li Z, Zhang X, Liu C, Ma J. Non-immune Cell Components in the Gastrointestinal Tumor Microenvironment Influencing Tumor Immunotherapy. Front Cell Dev Biol 2021; 9:729941. [PMID: 34722510 PMCID: PMC8549829 DOI: 10.3389/fcell.2021.729941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Interactions of genetic susceptibility factors, immune microenvironment, and microbial factors contribute to gastrointestinal tumorigenesis. The suppressive immune microenvironment reshaped by the tumors during gastrointestinal tumorigenesis directly contributes to T-cell depletion in tumor immunotherapy. Soluble factors secreted by tumor cells or stromal cells collectively shape the suppressive immune environment. Here, we reviewed the key factors in the gastrointestinal tumor microenvironment that influence tumor immunotherapy, focusing on the effects of fibroblasts, neuronal cells, soluble cytokines, exosomes, and the microbiome in tumor microenvironment. Research in this field has helped to identify more precise and effective biomarkers and therapeutic targets in the era of tumor immunotherapy.
Collapse
Affiliation(s)
- Zhengshuo Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Xiaoyue Zhang
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Can Liu
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| |
Collapse
|
135
|
Zheng Q, Duan L, Lou Y, Chao T, Guo G, Lu L, Zhang H, Zhao Y, Liang Y, Wang H. Slfn4 deficiency improves MAPK-mediated inflammation, oxidative stress, apoptosis and abates atherosclerosis progression in apolipoprotein E-deficient mice. Atherosclerosis 2021; 337:42-52. [PMID: 34757313 DOI: 10.1016/j.atherosclerosis.2021.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/12/2021] [Accepted: 10/15/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND AIMS Atherosclerosis, a progressive inflammatory disease characterized by elevated inflammation and lipid accumulation in the aortic endothelium, arises in part from the infiltration of inflammatory cells into the vascular wall. However, it is not fully defined how inflammatory cells, especially macrophages, affect the pathogenesis of atherosclerosis. Schlafen4 (Slfn4) mRNA is remarkably upregulated upon ox-LDL stimulation in macrophages. Nonetheless, the role of Slfn4 in foam cell formation remains unclear. METHODS To determine whether and how Slfn4 regulates lesion macrophage function during atherosclerosis,we engineered ApoE-/-Slfn4-/- double-deficient mice on an ApoE-/- background and evaluated the deficiency of Slfn4 expression in atherosclerotic lesion formation in vivo. RESULTS Our results demonstrate that total absence of SLFN4 and the bone marrow-restricted deletion of Slfn4 in ApoE-/- mice remarkably diminish inflammatory cell numbers within arterial plaques as well as limit development of atherosclerosis in moderate hypercholesterolemia condition. This is linked to a marked reduction in the expression of proinflammatory cytokines, the generation of the reactive oxygen species (ROS) and the apoptosis of cells. Furthermore, the activation of MAPKs and apoptosis signaling pathways is compromised in the absence of Slfn4. CONCLUSIONS These findings demonstrate a novel role of Slfn4 in modulating vascular inflammation and atherosclerosis, highlighting a new target for the related diseases.
Collapse
Affiliation(s)
- Qianqian Zheng
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Liangwei Duan
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Yunwei Lou
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Tianzhu Chao
- Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Guo Guo
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Liaoxun Lu
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China; Laboratory of Mouse Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Hongxia Zhang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Yucong Zhao
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China
| | - Yinming Liang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China.
| | - Hui Wang
- Henan Key Laboratory of Immunology and Targeted Drugs, Xinxiang Medical University, Xinxiang, People's Republic of China; Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, People's Republic of China.
| |
Collapse
|
136
|
Salemizadeh Parizi M, Salemizadeh Parizi F, Abdolhosseini S, Vanaei S, Manzouri A, Ebrahimzadeh F. Myeloid-derived suppressor cells (MDSCs) in brain cancer: challenges and therapeutic strategies. Inflammopharmacology 2021; 29:1613-1624. [PMID: 34613567 DOI: 10.1007/s10787-021-00878-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022]
Abstract
The most fatal malignancy of the central nervous system (CNS) is glioblastoma. Brain cancer is a 'cold' tumor because of fewer immunoregulatory cells and more immunosuppressive cells. Due to the cold nature of brain cancers, conventional treatments which are used to manage glioma patients show little effectiveness. Glioma patients even showed resistance to immune checkpoint blockade (ICB) and no significant efficacy. It has been shown that myeloid-derived suppressor cells (MDSCs) account for approximately 30-50% of the tumor mass in glioma. This study aimed to review MDSC function in brain cancer, as well as possible treatments and related challenges. In brain cancer and glioma, several differences in the context of MDSCs have been reported, including disagreements about the MDSC subtype that has the most inhibitory function in the brain, or inhibitory function of regulatory B cells (Bregs). There are also serious challenges in treating glioma patients. In addition to the cold nature of glioma, there are reports of an increase in MDSCs following conventional chemotherapy treatments. As a result, targeting MDSCs in combination with other therapies, such as ICB, is essential, and recent studies with the combination therapy approach have shown promising therapeutic effects in brain cancer.
Collapse
Affiliation(s)
| | | | | | - Shohreh Vanaei
- Department of Biomedical Engineering, Northeastern University, Boston, MA, USA
| | - Ali Manzouri
- School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Farnoosh Ebrahimzadeh
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
137
|
Li Y, He H, Jihu R, Zhou J, Zeng R, Yan H. Novel Characterization of Myeloid-Derived Suppressor Cells in Tumor Microenvironment. Front Cell Dev Biol 2021; 9:698532. [PMID: 34527668 PMCID: PMC8435631 DOI: 10.3389/fcell.2021.698532] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/09/2021] [Indexed: 11/21/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous group of cells generated in various pathologic conditions, which have been known to be key components of the tumor microenvironment (TME) involving in tumor immune tolerance. So MDSCs have been extensively researched recently. As its name suggests, immunosuppression is the widely accepted function of MDSCs. Aside from suppressing antitumor immune responses, MDSCs in the TME also stimulate tumor angiogenesis and metastasis, thereby promoting tumor growth and development. Therefore, altering the recruitment, expansion, activation, and immunosuppression of MDSCs could partially restore antitumor immunity. So, this view focused on the favorable TME conditions that promote the immunosuppressive effects of MDSCs and contribute to targeted therapies with increased precision for MDSCs.
Collapse
Affiliation(s)
- Yanan Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Hongdan He
- Immunotherapy Laboratory, Qinghai Tibet Plateau Research Institute, Southwest Minzu University, Chengdu, China
| | - Ribu Jihu
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Rui Zeng
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, China
| |
Collapse
|
138
|
Miller K, McVeigh CM, Barr EB, Herbert GW, Jacquez Q, Hunter R, Medina S, Lucas SN, Ali AMS, Campen MJ, Bolt AM. Inhalation of tungsten metal particulates alters the lung and bone microenvironments following acute exposure. Toxicol Sci 2021; 184:286-299. [PMID: 34498067 DOI: 10.1093/toxsci/kfab109] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inhalation of tungsten particulates is a relevant route of exposure in occupational and military settings. Exposure to tungsten alloys is associated with increased incidence of lung pathologies, including interstitial lung disease and cancer. We have demonstrated, oral exposure to soluble tungsten enhances breast cancer metastasis to the lungs through changes in the surrounding microenvironment. However, more research is required to investigate if changes in the lung microenvironment, following tungsten particulate exposure, can drive tumorigenesis or metastasis to the lung niche. This study examined if inhalation to environmentally relevant concentrations of tungsten particulates caused acute damage to the microenvironment in the lungs and/or systemically using a whole-body inhalation system. Twenty-four female BALB/c mice were exposed to Filtered Air, 0.60 mg/m3, or 1.7 mg/m3 tungsten particulates (< 1 µm) for 4 h. Tissue samples were collected at day 1 and 7 post-exposure. Tungsten accumulation in the lungs persisted up to 7 days post-exposure and produced acute changes to the lung microenvironment including increased macrophage and neutrophil infiltration, increased levels of pro-inflammatory cytokines IL-1β and CXCL1, and an increased percentage of activated fibroblasts (α-SMA+). Exposure to tungsten also resulted in systemic effects on the bone, including tungsten deposition and transient increases in gene expression of pro-inflammatory cytokines. Taken together, acute whole-body inhalation of tungsten particulates, at levels commonly observed in occupational and military settings, resulted in changes to the lung and bone microenvironments that may promote tumorigenesis or metastasis and be important molecular drivers of other tungsten-associated lung pathologies such as interstitial lung disease.
Collapse
Affiliation(s)
- Kara Miller
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Charlotte M McVeigh
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Edward B Barr
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Guy W Herbert
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Quiteria Jacquez
- College of Nursing, University of New Mexico, Albuquerque, NM, 87131
| | - Russell Hunter
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Sebastian Medina
- Department of Biology, New Mexico Highlands University, Las Vegas, NM, 87701
| | - Selita N Lucas
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Abdul-Mehdi S Ali
- Department of Earth and Planetary Sciences, University of New Mexico, Albuquerque, NM, 87131
| | - Matthew J Campen
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| | - Alicia M Bolt
- College of Pharmacy, Department of Pharmaceutical Sciences, The University of New Mexico, Albuquerque, NM 87131
| |
Collapse
|
139
|
Helicobacter pylori-Induced Inflammation: Possible Factors Modulating the Risk of Gastric Cancer. Pathogens 2021; 10:pathogens10091099. [PMID: 34578132 PMCID: PMC8467880 DOI: 10.3390/pathogens10091099] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/21/2021] [Accepted: 08/25/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic inflammation and long-term tissue injury are related to many malignancies, including gastric cancer (GC). Helicobacter pylori (H. pylori), classified as a class I carcinogen, induces chronic superficial gastritis followed by gastric carcinogenesis. Despite a high prevalence of H. pylori infection, only about 1–3% of people infected with this bacterium develop GC worldwide. Furthermore, the development of chronic gastritis in some, but not all, H. pylori-infected subjects remains unexplained. These conflicting findings indicate that clinical outcomes of aggressive inflammation (atrophic gastritis) to gastric carcinogenesis are influenced by several other factors (in addition to H. pylori infection), such as gut microbiota, co-existence of intestinal helminths, dietary habits, and host genetic factors. This review has five goals: (1) to assess our current understanding of the process of H. pylori-triggered inflammation and gastric precursor lesions; (2) to present a hypothesis on risk modulation by the gut microbiota and infestation with intestinal helminths; (3) to identify the dietary behavior of the people at risk of GC; (4) to check the inflammation-related genetic polymorphisms and role of exosomes together with other factors as initiators of precancerous lesions and gastric carcinoma; and (5) finally, to conclude and suggest a new direction for future research.
Collapse
|
140
|
Chai S, Guo S, Yang J, Lu S, Yue Y, Li H, Sun P, Zhang T, Xu B, Sun H, Si S, Zhou J, Yang J, Yang H, Li Z, Cui Y. Inflammation is involved in response of gastric mucosal epithelial cells under simulated microgravity by integrated transcriptomic analysis. Am J Transl Res 2021; 13:9195-9207. [PMID: 34540035 PMCID: PMC8430122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 05/26/2021] [Indexed: 06/13/2023]
Abstract
Astronauts suffer from inflammatory changes induced by microgravity during space flight. Microgravity can significantly affect the inflammatory response of various cell types and multiple systems of the human body, such as cardiovascular system, skeletal muscle system, and digestive system. The aim of this research was to identify the key genes and pathways of gastric mucosa affected by microgravity. Human gastric mucosal epithelial GES-1 cells were cultured in a rotary cell culture system (RCCS) bioreactor to simulate microgravity. The gene expression profiles of GES-1 cells were obtained using Illumina sequencing platform and differentially expressed genes were identified by DESeq2 software, then Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed. Subsequently, a protein-protein interaction (PPI) network was constructed. Compared with a normal gravity (NG) group, a total of 943 DEGs, including 192 downregulated genes and 751 upregulated genes, were identified. These DEGs were associated with findings that included response to interleukin-1, positive regulation of inflammatory response, and positive regulation of neuroinflammatory response. Furthermore, these DEGs were mainly enriched in herpes simplex virus 1 infection, cytokine-cytokine receptor interaction, and NOD-like receptor signaling pathway. Thus, 21 hub genes were identified from PPI network, including IL6, IL1B, ITGAM, CXCL8, ITGAX, CCL5, SERPINA1, APOE, CSF1R, VWF, GBP1, APOB, CYBB, HLA-DRB1, CD68, FGG, FGA, OASL, NOD2, OAS2 and FCGR2A. These findings suggested that simulated microgravity upregulated inflammation-related genes and pathways of GES-1 cells, which may play important roles in the response to microgravity and provide useful information for preventing mucosal damage in astronauts. In conclusion, this study revealed the key genes and pathways associated with simulated microgravity and indicated that simulated microgravity induced an inflammatory response in gastric mucosal epithelial cells.
Collapse
Affiliation(s)
- Shaobin Chai
- Medical School of Chinese PLA Beijing 100853, China
| | - Song Guo
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Jiaqi Yang
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Shengyu Lu
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Yuan Yue
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Hao Li
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Peiming Sun
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Tao Zhang
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Binxin Xu
- Special Medical Laboratory Center, Strategic Support Force Medical Center Beijing 100101, China
| | - Hongwei Sun
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Shaoyan Si
- Special Medical Laboratory Center, Strategic Support Force Medical Center Beijing 100101, China
| | - Jinlian Zhou
- Department of Pathology, Strategic Support Force Medical Center Beijing 100101, China
| | - Jianwu Yang
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Heming Yang
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| | - Zhengpeng Li
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital Beijing 100853, China
| | - Yan Cui
- Medical School of Chinese PLA Beijing 100853, China
- Department of General Surgery, Strategic Support Force Medical Center Beijing 100101, China
| |
Collapse
|
141
|
Tang D, Huang W, Yang Z, Wu X, Sang X, Wang K, Cao G. Identification and validation of 12 immune-related genes as a prognostic signature for colon adenocarcinoma. J Biochem Mol Toxicol 2021; 35:e22852. [PMID: 34396630 DOI: 10.1002/jbt.22852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 03/30/2021] [Accepted: 07/14/2021] [Indexed: 11/10/2022]
Abstract
Colon adenocarcinoma (COAD) is a common malignant tumor of the digestive tract that threatens human health seriously. Thus, it is urgent to explore biomarkers that can be used to evaluate a patient's survival prognosis overall as a supplementary treatment. RNA-seq expression profiles were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus, and Lasso and multivariate Cox regression analyses were used for developing the prognostic model. Finally, a nomogram comprising the prognostic model was established to evaluate survival overall. A risk model comprised of a total of 12 immune-related gene pairs was constructed. Further analysis revealed the model's independent prognostic ability in relation to other clinical characteristics. This model's nomogram could help clinicians choose personalized treatment for COAD patients. This model has significant potential to complement COAD's clinical identifying characteristics, and also provide new insights into the identification of colon cancer patients with a high risk of death.
Collapse
Affiliation(s)
- Dongxin Tang
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Wei Huang
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Zhu Yang
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kuilong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
142
|
Busada JT, Khadka S, Peterson KN, Druffner SR, Stumpo DJ, Zhou L, Oakley RH, Cidlowski JA, Blackshear PJ. Tristetraprolin Prevents Gastric Metaplasia in Mice by Suppressing Pathogenic Inflammation. Cell Mol Gastroenterol Hepatol 2021; 12:1831-1845. [PMID: 34358715 PMCID: PMC8554534 DOI: 10.1016/j.jcmgh.2021.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 07/27/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Aberrant immune activation is associated with numerous inflammatory and autoimmune diseases and contributes to cancer development and progression. Within the stomach, inflammation drives a well-established sequence from gastritis to metaplasia, eventually resulting in adenocarcinoma. Unfortunately, the processes that regulate gastric inflammation and prevent carcinogenesis remain unknown. Tristetraprolin (TTP) is an RNA-binding protein that promotes the turnover of numerous proinflammatory and oncogenic messenger RNAs. Here, we assess the role of TTP in regulating gastric inflammation and spasmolytic polypeptide-expressing metaplasia (SPEM) development. METHODS We used a TTP-overexpressing model, the TTPΔadenylate-uridylate rich element mouse, to examine whether TTP can protect the stomach from adrenalectomy (ADX)-induced gastric inflammation and SPEM. RESULTS We found that TTPΔadenylate-uridylate rich element mice were completely protected from ADX-induced gastric inflammation and SPEM. RNA sequencing 5 days after ADX showed that TTP overexpression suppressed the expression of genes associated with the innate immune response. Importantly, TTP overexpression did not protect from high-dose-tamoxifen-induced SPEM development, suggesting that protection in the ADX model is achieved primarily by suppressing inflammation. Finally, we show that protection from gastric inflammation was only partially due to the suppression of Tnf, a well-known TTP target. CONCLUSIONS Our results show that TTP exerts broad anti-inflammatory effects in the stomach and suggest that therapies that increase TTP expression may be effective treatments of proneoplastic gastric inflammation. Transcript profiling: GSE164349.
Collapse
Affiliation(s)
- Jonathan T. Busada
- Molecular Endocrinology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina,Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, West Virginia,Correspondence Address correspondence to: Jonathan T. Busada, PhD, Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 64 Medical Center Drive, PO Box 9177, Morgantown, West Virginia 26506.
| | - Stuti Khadka
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Kylie N. Peterson
- Molecular Endocrinology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Sara R. Druffner
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Deborah J. Stumpo
- Post-Transcriptional Gene Expression Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Lecong Zhou
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Robert H. Oakley
- Molecular Endocrinology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - John A. Cidlowski
- Molecular Endocrinology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Perry J. Blackshear
- Post-Transcriptional Gene Expression Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| |
Collapse
|
143
|
Briukhovetska D, Dörr J, Endres S, Libby P, Dinarello CA, Kobold S. Interleukins in cancer: from biology to therapy. Nat Rev Cancer 2021; 21:481-499. [PMID: 34083781 PMCID: PMC8173513 DOI: 10.1038/s41568-021-00363-z] [Citation(s) in RCA: 385] [Impact Index Per Article: 96.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
Interleukins and associated cytokines serve as the means of communication for innate and adaptive immune cells as well as non-immune cells and tissues. Thus, interleukins have a critical role in cancer development, progression and control. Interleukins can nurture an environment enabling and favouring cancer growth while simultaneously being essential for a productive tumour-directed immune response. These properties of interleukins can be exploited to improve immunotherapies to promote effectiveness as well as to limit side effects. This Review aims to unravel some of these complex interactions.
Collapse
Affiliation(s)
- Daria Briukhovetska
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Janina Dörr
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany
- German Center for Translational Cancer Research (DKTK), Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU, Munich, Germany.
- German Center for Translational Cancer Research (DKTK), Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| |
Collapse
|
144
|
Chen H, Jiang T, Lin F, Guan H, Zheng J, Liu Q, Du B, Huang Y, Lin X. PD-1 inhibitor combined with apatinib modulate the tumor microenvironment and potentiate anti-tumor effect in mice bearing gastric cancer. Int Immunopharmacol 2021; 99:107929. [PMID: 34343940 DOI: 10.1016/j.intimp.2021.107929] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE To explore the effect of programmed death 1 (PD-1) inhibitor combined with apatinib on immune regulation and efficacy of the combined therapy in mice bearing gastric cancer (MBGC), and to provide a research basis for enhancing the benefit of immunotherapy in advanced gastric cancer (AGC). METHODS MBGC were divided into normal saline group (group NS), apatinib group (group A), PD-1 inhibitors group (group B) and PD-1 inhibitors combined with apatinib group (group C). Tumor inhibition rates were calculated. Cytokine levels and expression of immune cells and molecules were detected, and the pathological manifestations of tumor tissues were observed. RESULTS Group C had the smallest tumor volume (115.17 ± 16.08 mm3) with a tumor inhibition rate of 89.4% ± 0.69%, significantly increased levels of CD4+T and CD8+T cells in tumor tissues (P < 0.01), the down-regulated proportion of myeloid-derived suppressor cells (MDSCs) (P < 0.01), and levels of PD-1 of CD8+T cells (PD-1+CD8+T) (P < 0.01). There was no difference in the levels of PD-1+CD8+T, CD4+T cells, and MDSCs between groups B and C. Besides, combination therapy increased the levels of interleukin-2 (IL-2), interferon-gamma (IFN-γ), and tumor necrosis factor-ɑ (TNF-ɑ) in tumor tissue and serum. We also found that the anti-angiogenic effect of apatinib increased programmed death ligand-1 (PD-L1) levels, down-regulated vascular endothelial growth factor receptor 2 (VEGFR-2) levels, and induced an increase in the extent of tumor tissue necrosis. CONCLUSION PD-1 inhibitors in combination with apatinib may help improve treatment outcomes and increase survival benefits in patients with AGC.
Collapse
Affiliation(s)
- Hao Chen
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Tao Jiang
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Fangyu Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Hongdan Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Jianwei Zheng
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Qing Liu
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Bing Du
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Yeyuan Huang
- Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
| | - Xiaoyan Lin
- Department of Medical Oncology, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, People's Republic of China.
| |
Collapse
|
145
|
NF-κB in Gastric Cancer Development and Therapy. Biomedicines 2021; 9:biomedicines9080870. [PMID: 34440074 PMCID: PMC8389569 DOI: 10.3390/biomedicines9080870] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/09/2021] [Accepted: 07/20/2021] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer is considered one of the most common causes of cancer-related death worldwide and, thus, a major health problem. A variety of environmental factors including physical and chemical noxae, as well as pathogen infections could contribute to the development of gastric cancer. The transcription factor nuclear factor kappa B (NF-κB) and its dysregulation has a major impact on gastric carcinogenesis due to the regulation of cytokines/chemokines, growth factors, anti-apoptotic factors, cell cycle regulators, and metalloproteinases. Changes in NF-κB signaling are directed by genetic alterations in the transcription factors themselves, but also in NF-κB signaling molecules. NF-κB actively participates in the crosstalk of the cells in the tumor micromilieu with divergent effects on the heterogeneous tumor cell and immune cell populations. Thus, the benefits/consequences of therapeutic targeting of NF-κB have to be carefully evaluated. In this review, we address recent knowledge about the mechanisms and consequences of NF-κB dysregulation in gastric cancer development and therapy.
Collapse
|
146
|
Jiang L, Ye WC, Li Z, Yang Y, Dai W, Li M. Anticancer effects of dihydromyricetin on the proliferation, migration, apoptosis and in vivo tumorigenicity of human hepatocellular carcinoma Hep3B cells. BMC Complement Med Ther 2021; 21:194. [PMID: 34229692 PMCID: PMC8258952 DOI: 10.1186/s12906-021-03356-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) represents a serious public health problem worldwide and has high morbidity and mortality. Dihydromyricetin (DHM) exhibits anticancer effect on a variety of malignancies, but its anticancer function of DHM in HCC has been unclear. The aim of this study was designed to investigate the anticancer effect of DHM on cell apoptosis, proliferation, migration and invasion of hepatoma carcinoma cells. Methods Cultured Hep3B cells were treated with different DHM concentrations, followed by cell apoptosis, proliferation, migration and invasion were examined by CCK-8, colony formation assay, wound healing, Transwell and flow cytometry, respectively. The mRNA and protein expression of BCL-2, Cleaved-caspase 3, Cleaved-caspase 9, BAK, BAX and BAD were validated by western blot. Results DHM markedly suppressed proliferation, migration, invasion and facilitated apoptosis in Hep3B cells. Mechanistically, DHM significantly downregulated the Bcl-2 expression, and upregulated the mRNA and protein levels of Cleaved-Caspase 3, Cleaved- Caspase 9, Bak, Bax and Bad. Furthermore, in the nude mice tumorigenic model, DHM treatment greatly decreased the weight of the HCC cancers compared to the weights in control and NDP group. Conclusions DHM could suppress cell proliferation, migration, invasion, and facilitated apoptosis in Hep3B cells. These findings could provide novel insights to develop potential therapeutic strategy for the clinical treatment of HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03356-5.
Collapse
Affiliation(s)
- Lianggui Jiang
- Laboratory of Hepatobiliary Surgery, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China.,Department of Thyroid and Breast Surgery, The People's Hospital of Ganzhou, Ganzhou Affiliated Hospital of Nanchang University, Ganzhou, Jiangxi, 341000, P.R. China
| | - Wen-Chu Ye
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, 511518, Guangdong, China
| | - Zuobiao Li
- Laboratory of Hepatobiliary Surgery, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Yongguang Yang
- Laboratory of Hepatobiliary Surgery, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Wei Dai
- Laboratory of Hepatobiliary Surgery, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China
| | - Mingyi Li
- Laboratory of Hepatobiliary Surgery, Zhanjiang Key Laboratory of Hepatobiliary Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, People's Republic of China.
| |
Collapse
|
147
|
Groth C, Weber R, Lasser S, Özbay FG, Kurzay A, Petrova V, Altevogt P, Utikal J, Umansky V. Tumor promoting capacity of polymorphonuclear myeloid-derived suppressor cells and their neutralization. Int J Cancer 2021; 149:1628-1638. [PMID: 34224592 DOI: 10.1002/ijc.33731] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 12/17/2022]
Abstract
Myeloid-derived suppressor cells (MDSC) represent a highly immunosuppressive population that expands in tumor bearing hosts and inhibits both T and NK cell antitumor effector functions. Among MDSC subpopulations, the polymorphonuclear (PMN) one is gaining increasing interest since it is a predominant MDSC subset in most cancer entities and inherits unique properties to facilitate metastatic spread. In addition, further improvement in distinguishing PMN-MDSC from neutrophils has contributed to the design of novel therapeutic approaches. In this review, we summarize the current view on the origin of PMN-MDSC and their relation to classical neutrophils. Furthermore, we outline the metastasis promoting features of these cells and promising strategies of their targeting to improve the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Christopher Groth
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department for Immunobiochemistry, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rebekka Weber
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Samantha Lasser
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Feyza Gül Özbay
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Annina Kurzay
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Faculty of Biosciences, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany
| | - Vera Petrova
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Mannheim Institute for Innate Immunoscience (MI3), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
148
|
Abstract
Helicobacter pylori is present in approximately one-half of the world's population. There are significant differences in prevalence based on region, age, race/ethnicity, and socioeconomic status. H pylori is the most common cause of infection-related cancers. Studies have demonstrated the relationship between H pylori infection and gastric adenocarcinoma and mucosa-associated lymphoid tissue lymphoma. H pylori has features and enzymatic properties allowing it to survive in the acidic stomach environment, and has specific virulence factors that promote an increased risk of gastric pathology. Eradication of H pylori is first-line therapy for mucosa-associated lymphoid tissue lymphoma and decreases the risk of gastric adenocarcinoma.
Collapse
|
149
|
Yee PP, Li W. Tumor necrosis: A synergistic consequence of metabolic stress and inflammation. Bioessays 2021; 43:e2100029. [PMID: 33998010 PMCID: PMC8217290 DOI: 10.1002/bies.202100029] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/01/2021] [Accepted: 04/07/2021] [Indexed: 12/14/2022]
Abstract
Tumor necrosis is a common histological feature and poor prognostic predictor in various cancers. Despite its significant clinical implications, the mechanism underlying tumor necrosis remains largely unclear due to lack of appropriate pre-clinical modeling. We propose that tumor necrosis is a synergistic consequence of metabolic stress and inflammation, which lead to oxidative stress-induced cell death, such as ferroptosis. As a natural consequence of tumor expansion, tumor cells are inevitably stripped of vascular supply, resulting in deprivation of oxygen and nutrients. The resulting metabolic stress has commonly been considered the cause of tumor necrosis. Recent studies found that immune cells, such as neutrophils, when recruited to tumors, can directly trigger ferroptosis in tumor cells, suggesting that immune cells can be involved in amplifying tumor necrosis. This article will discuss potential mechanisms underlying tumor necrosis development and its impact on tumor progression as well as the immune response to tumors.
Collapse
Affiliation(s)
- Patricia P. Yee
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Medical Scientist Training Program, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
150
|
Zhong C, Wang R, Hua M, Zhang C, Han F, Xu M, Yang X, Li G, Hu X, Sun T, Ji C, Ma D. NLRP3 Inflammasome Promotes the Progression of Acute Myeloid Leukemia via IL-1β Pathway. Front Immunol 2021; 12:661939. [PMID: 34211462 PMCID: PMC8239362 DOI: 10.3389/fimmu.2021.661939] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/17/2021] [Indexed: 01/18/2023] Open
Abstract
NLRP3 inflammasome has been reported to be associated with the pathogenesis of multiple solid tumors. However, the role of NLRP3 inflammasome in acute myeloid leukemia (AML) remains unclear. We showed that NLRP3 inflammasome is over-expressed and highly activated in AML bone marrow leukemia cells, which is correlated with poor prognosis. The activation of NLRP3 inflammasome in AML cells promotes leukemia cells proliferation, inhibits apoptosis and increases resistance to chemotherapy, while inactivation of NLRP3 by caspase-1 or NF-κB inhibitor shows leukemia-suppressing effects. Bayesian networks analysis and cell co-culture tests further suggest that NLRP3 inflammasome acts through IL-1β but not IL-18 in AML. Knocking down endogenous IL-1β or anti-IL-1β antibody inhibits leukemia cells whereas IL-1β cytokine enhances leukemia proliferation. In AML murine model, up-regulation of NLRP3 increases the leukemia burden in bone marrow, spleen and liver, and shortens the survival time; furthermore, knocking out NLRP3 inhibits leukemia progression. Collectively, all these evidences demonstrate that NLRP3 inflammasome promotes AML progression in an IL-1β dependent manner, and targeting NLRP3 inflammasome may provide a novel therapeutic option for AML.
Collapse
Affiliation(s)
- Chaoqing Zhong
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China.,Department of Hematology, Shandong Yantai Mountain Hospital, Yantai, China
| | - Ruiqing Wang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Mingqiang Hua
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China.,Department of Emergency, Qilu Hospital of Shandong University, Jinan, China
| | - Chen Zhang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Fengjiao Han
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Miao Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Xinyu Yang
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiang Hu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Tao Sun
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Chunyan Ji
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|