151
|
Wang Y, You K, You Y, Li Q, Feng G, Ni J, Cao X, Zhang X, Wang Y, Bao W, Wang X, Chen T, Li H, Huang Y, Lyu J, Yu S, Li H, Xu S, Zeng K, Shen X. Paeoniflorin prevents aberrant proliferation and differentiation of intestinal stem cells by controlling C1q release from macrophages in chronic colitis. Pharmacol Res 2022; 182:106309. [PMID: 35716915 DOI: 10.1016/j.phrs.2022.106309] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022]
Abstract
The pathological features of inflammatory bowel disease necessitate therapeutic strategies aimed at restoring intestinal mucosal barrier function in addition to controlling inflammation. Paeoniflorin, a bioactive herbal constituent isolated from the root of Paeonia albiflora Pall, has been reported to protect against acute colitis in mice. However, the direct molecular target of paeoniflorin in preventing colitis remains elusive. Here, we evaluated the therapeutical effects of Paeoniflorin using IL-10-/- chronic colitis model, and explored the precise mechanism of action involved. Our results demonstrated that intragastric administration of Paeoniflorin significantly ameliorated inflammatory response and restored the aberrant intestinal proliferation and differentiation in IL-10-/-colitis mice. By utilizing a chemical biology approach, we identified C1qa, a crucial component of C1q, is the direct target of Paeoniflorin. Binding of Paeoniflorin to C1qa prevented the cleavage of C1q on macrophages, resulting in the aggregation of surface membrane-anchored C1q and the diminished C1q secretion. The excessive surface membrane-anchored C1q significantly enhanced the phagocytic capability of macrophages and promoted the elimination of infiltrated bacteria and inflammatory cells in mouse colon. The reduced C1q secretion conferred by Paeoniflorin dampened Wnt/β-catenin signaling activation, thereby rectifying the aberrant proliferation and differentiation of intestinal stem cells (ISCs). In summary, our study demonstrates that Paeoniflorin can orchestrate mucosal healing and intestinal inflammation elimination through C1q-bridged macrophage-ISCs crosstalk, highlighting a novel strategy to treat chronic colitis by restoring mucosal homeostasis via targeting C1q.
Collapse
Affiliation(s)
- Yirui Wang
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Keyuan You
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yan You
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Qian Li
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Guize Feng
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiahui Ni
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xinyue Cao
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xiaowen Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yanhang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Weilian Bao
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Xu Wang
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Tongqing Chen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haidong Li
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yuran Huang
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaren Lyu
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shihang Yu
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hong Li
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Suowen Xu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China.
| | - Xiaoyan Shen
- Department of Pharmacology & the Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai 201203, China.
| |
Collapse
|
152
|
Zizhu Ointment Accelerates Wound-Healing of Diabetic Ulcers through Promoting M2 Macrophage Polarization via Downregulating the Notch4 Signaling Pathway. COMPUTATIONAL INTELLIGENCE AND NEUROSCIENCE 2022; 2022:5173836. [PMID: 35619768 PMCID: PMC9129934 DOI: 10.1155/2022/5173836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 11/17/2022]
Abstract
Objective The long-term clinical practice shows that Zizhu ointment (ZZO) is an empirical formula for the treatment of diabetic ulcers (DUs). In this study, we investigated the underlying mechanism of ZZO in the treatment of DU mice. Methods Through streptozotocin induction and high-fat diet, a DU mouse model was established and ZZO was given for treatment. The activation of Notch4 signaling was examined by immunofluorescence staining, RT-PCR, as well as Western blotting. Flow cytometry was performed to detect the counts of F4/80+ cells, M1 and M2 macrophages, as well as the expression of CD11c, CD206, etc. The role of Notch4 in wound healing in diabetic mice was verified by Notch4 inhibitors and agonists. Results Accelerated wound healing and decreased expression levels of Notch4 and its target genes and ligands were observed in diabetic mice treated with ZZO. ZZO promoted M2 macrophage polarization, downregulated the expression of proinflammatory factors, and upregulated the levels of anti-inflammatory factors. The same tendency was observed in diabetic mice after treatment with Notch4 inhibitors. Knockout of Notch4 accelerated the wound healing rate as well. Conclusions ZZO accelerates wound healing of diabetic mice through inhibiting the activation of Notch4 signaling, promoting M2 macrophage polarization, and alleviating inflammation.
Collapse
|
153
|
Qiu J, Ma Y, Qiu J. Regulation of intestinal immunity by dietary fatty acids. Mucosal Immunol 2022; 15:846-856. [PMID: 35821290 DOI: 10.1038/s41385-022-00547-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/28/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023]
Abstract
Dietary fatty acids are absorbed through the intestine and are fundamental for cellular energy provision and structural formation. Dietary fatty acids profoundly affect intestinal immunity and influence the development and progression of inflammatory bowel disease, intestinal infections and tumors. Although different types of fatty acids exert differential roles in intestinal immunity, a western diet, rich in saturated fatty acids with abundant carbohydrates and studied as high-fat diet (HFD) in animal experiments, disturbs intestinal homeostasis and plays a pathogenic role in intestinal inflammatory diseases. Here, we review recent findings on the regulation of intestinal immunity by dietary fatty acids, focusing on HFD. We summarize HFD-altered immune responses leading to susceptibility to intestinal pathology and dissect the mechanisms involving the impact of HFD on immune cells, intestinal epithelial cells and the microbiota. Understanding the perturbation of intestinal immunity by HFD will provide new strategies for prevention and treatment of intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Jinxin Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanhui Ma
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
154
|
Pang C, Wen C, Liang Y, Luo H, Wei L, Liu H, Qin T, Tan H, He C, Liu Y, Chen Y, Zeng S, Zhou C. Asperosaponin VI protects mice from sepsis by regulating Hippo and Rho signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 99:154010. [PMID: 35228043 DOI: 10.1016/j.phymed.2022.154010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 01/09/2022] [Accepted: 02/16/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE To explore the novel protective effect of Asperosaponin VI (AVI) on sepsis and its potential mechanism. METHODS In in vitro experiments, bone marrow mononuclear cells and THP-1-derived cells were used to evaluate the viability of AVI treatment. Besides, the quantitative real-time PCR and Western blot were adopted to explore the protective effect of AVI on LPS-induced inflammation. For in vivo work, the effect of AVI on mice was evaluated by using both CLP-induced and the LPS-induced sepsis mice model. The fluctuation of anal temperature and the behavior of mice were recorded after surgery. Further, the content of bacteria in peritoneal lavage fluid was detected, as well as the levels of ALT, AST, LD and LDH in serum with ELISA. H&E staining and real-time PCR were used to evaluate the histopathology of liver, spleen and lung. Finally, relevant signaling pathways were detected by Western blot, real-time PCR and immunohistochemistry. RESULTS AVI inhibited the expression of inflammatory factors in both CLP-induced and LPS-induced sepsis mice models, and reduced the number of bacteria in abdominal lavage fluid. The preventive treatment with AVI alleviated sepsis-induced organ injuries, reduced inflammatory responses, which was through inhibiting Hippo and Rho signaling pathway. CONCLUSIONS This study indicated that AVI effectively protected mice from sepsis by down-regulating the activation of Hippo signaling and Rho family, and reducing inflammation and organ damage. However, conventional treatment was using antibiotics, and its mechanism was different with AVI.
Collapse
Affiliation(s)
- Caixia Pang
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Cailing Wen
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Yanxiang Liang
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Hui Luo
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Linlin Wei
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China; Deparment of Pharmacy, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou 510317, China
| | - Haiqian Liu
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Tian Qin
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Huijing Tan
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Chonghua He
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China
| | - Ying Liu
- School of Pharmacy, Guangzhou Xinhua University, Guangzhou 510520, China
| | - Yang Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Siyu Zeng
- Deparment of Pharmacy, Guangdong Second Provincial General Hospital, No. 466 Xingang Middle Road, Haizhu District, Guangzhou 510317, China.
| | - Chun Zhou
- SMU-KI United Medical Inflammatory Center, Guangdong Provincial Key Laboratory of Shock and Microcirculation, School of Pharmaceutical Sciences, Southern Medical University, No. 1023-1063, Shatai South Road, Baiyun District, Guangzhou 510515, China.
| |
Collapse
|
155
|
Sung PS, Park DJ, Roh PR, Mun KD, Cho SW, Lee GW, Jung ES, Lee SH, Jang JW, Bae SH, Choi JY, Choi J, Ahn J, Yoon SK. Intrahepatic inflammatory IgA +PD-L1 high monocytes in hepatocellular carcinoma development and immunotherapy. J Immunother Cancer 2022; 10:e003618. [PMID: 35577505 PMCID: PMC9114848 DOI: 10.1136/jitc-2021-003618] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND IgA neutralizes pathogens to prevent infection at mucosal sites. However, emerging evidence shows that IgA contributes to aggravating inflammation or dismantling antitumor immunity in human diseased liver. The aim of this study was to elucidate the roles of inflammation-induced intrahepatic inflammatory IgA+ monocytes in the development of hepatocellular carcinoma (HCC). METHODS Patient cohorts including steatohepatitis cohort (n=61) and HCC cohort (n=271) were established. Patients' surgical and biopsy specimens were analyzed using immunohistochemistry. Multicolor flow cytometry was performed with a subset of patient samples. Single-cell RNA-Seq analysis was performed using Gene Expression Omnibus (GEO) datasets. Additionally, we performed in vitro differentiation of macrophages, stimulation with coated IgA, and RNA sequencing. Hepa1-6 cells and C57BL/6N mice were used to obtain HCC syngeneic mouse models. RESULTS Serum IgA levels were associated (p<0.001) with fibrosis progression and HCC development in patients with chronic liver diseases. Additionally, immunohistochemical staining of inflamed livers or HCC revealed IgA positivity in monocytes, with a correlation between IgA+ cell frequency and IgA serum levels. Compared with IgA- monocytes, intrahepatic IgA+ monocytes expressed higher levels of programmed death-ligand 1 (PD-L1) in inflamed livers and in HCC tumor microenvironment. Single-cell RNA sequencing using NCBI GEO database indicated an upregulation in inflammation-associated genes in the monocytes of patients whose plasma cell IGHA1 expression was greater than or equal to the median value. Bulk RNA sequencing demonstrated that in vitro stimulation of M2-polarized macrophages using coated IgA complex induced PD-L1 upregulation via YAP-mediated signaling. In vivo blockade of IgA signaling decreased the number of tumor-infiltrating IgA+PD-L1high macrophages and increased the number of CD69+CD8+ T cells to enhance antitumor effects in HCC mice models. CONCLUSIONS Overall, the findings of this study showed that serum IgA levels was correlated with intrahepatic and intratumoral infiltration of inflammatory IgA+PD-L1high monocytes in chronic liver diseases and HCC, providing potential therapeutic targets.
Collapse
Affiliation(s)
- Pil Soo Sung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Dong Jun Park
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Pu Reun Roh
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Kyoung Do Mun
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Sung Woo Cho
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Gil Won Lee
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| | - Eun Sun Jung
- Department of Hospital Pathology, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jeong Won Jang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Si Hyun Bae
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Eunpyeong St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jong Young Choi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Jonghwan Choi
- Department of Computer Science, Yonsei University, Seoul, South Korea
| | - Jaegyoon Ahn
- Department of Computer Science & Engineering, Incheon National University, Incheon, South Korea
| | - Seung Kew Yoon
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
- The Catholic University Liver Research Center, College of Medicine, Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
156
|
Liu XY, Zhou K, Tian KJ, Yan BJ, Ren Z, Zhou ZX, Xiong WH, Jiang ZS. Hippo: a new hub for atherosclerotic disease. Curr Pharm Des 2022; 28:1321-1328. [DOI: 10.2174/1381612828666220428090540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/15/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
Hippo,an evolutionarily conserved kinase cascade reaction in organisms,can respond to a set of signals,such as mechanical signals and cell metabolism,to maintain cell growth,differentiation,tissue/organ development and homeostasis.In the past ten years,HIPPO has controlled the development of tissues and organs by regulating the process of cell proliferation,especially in the field of cardiac regeneration after myocardial infarction.This suggests that HIPPO signaling is closely linked to cardiovascular disease.Atherosclerosis is the most common disease of the cardiovascular system. It is characterised by chronic inflammation of the vascular wall, mainly involving dysfunction of endothelial cells, smooth muscle cells and macrophages.Oxidized Low density lipoprotein (LDL) damages the barrier function of endothelial cells, which enter the middle membrane of the vascular wall, accelerates the formation of foam cells and promotes the occurrence and development of atherosclerosis.Autophagy is associated with the development of atherosclerosis.However, the mechanism of HIPPO regulation of atherosclerosis has not meant to clarified.In view of the pivotal role of this signaling pathway in maintaining cell growth,proliferation and differentiation,the imbalance of Hippo is related to atherosclerosis and related diseases.In this review,we emphasized Hippo as a hub for regulating atherosclerosis and discussed its potential targets in pathophysiology,human diseases,and related pharmacology.
Collapse
Affiliation(s)
- Xi-Yan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Kun Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Kai-Jiang Tian
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Bin-Jie Yan
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhi-Xiang Zhou
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Wen-Hao Xiong
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, International Joint Laboratory for Arteriosclerotic Disease Research of Hunan Province, University of South China, Hengyang, China, 421001
| |
Collapse
|
157
|
Wang Y, Shi R, Zhai R, Yang S, Peng T, Zheng F, Shen Y, Li M, Li L. Matrix stiffness regulates macrophage polarization in atherosclerosis. Pharmacol Res 2022; 179:106236. [PMID: 35483516 DOI: 10.1016/j.phrs.2022.106236] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/01/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and the pathological basis of many fatal cardiovascular diseases. Macrophages, the main inflammatory cells in atherosclerotic plaque, have a paradox role in disease progression. In response to different microenvironments, macrophages mainly have two polarized directions: pro-inflammatory macrophages and anti-inflammatory macrophages. More and more evidence shows that macrophage is mechanosensitive and matrix stiffness regulate macrophage phenotypes in atherosclerosis. However, the molecular mechanism of matrix stiffness regulating macrophage polarization still lacks in-depth research, which hinders the development of new anti-atherosclerotic therapies. In this review, we discuss the important role of matrix stiffness in regulating macrophage polarization through mechanical signal transduction (Hippo, Piezo, cytoskeleton, and integrin) and epigenetic mechanisms (miRNA, DNA methylation, and histone). We hope to provide a new perspective for atherosclerosis therapy by targeting matrix stiffness and macrophage polarization.
Collapse
Affiliation(s)
- Yin Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Ruotong Shi
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Ran Zhai
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Shiyan Yang
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Tianqi Peng
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - Fuwen Zheng
- Norman Bethune College of Medicine, Jilin University, Changchun 130021, China
| | - YanNan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130021, China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
158
|
Huang C, Wang J, Liu H, Huang R, Yan X, Song M, Tan G, Zhi F. Ketone body β-hydroxybutyrate ameliorates colitis by promoting M2 macrophage polarization through the STAT6-dependent signaling pathway. BMC Med 2022; 20:148. [PMID: 35422042 PMCID: PMC9011974 DOI: 10.1186/s12916-022-02352-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/24/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Ketone body β-hydroxybutyrate (BHB) has received more and more attentions, because it possesses a lot of beneficial, life-preserving effects in the fields of clinical science and medicine. However, the role of BHB in intestinal inflammation has not yet been investigated. METHODS Colonic mucosa of inflammatory bowel disease (IBD) patients and healthy controls were collected for evaluation of BHB level. Besides, the therapeutic effect of exogenous BHB in a murine model of acute dextran sulfate sodium (DSS)-induced colitis were assessed by body weight change, colon length, disease activity index, and histopathological sections. The regulatory effectors of BHB were analyzed by RT-qPCR, immunofluorescence, and microbe analysis in vivo. Moreover, the molecular mechanism of BHB was further verified in bone marrow-derived macrophages (BMDMs). RESULTS In this study, significantly reduced BHB levels were found in the colonic mucosa from IBD patients and correlated with IBD activity index. In addition, we demonstrated that the administration of exogenous BHB alleviated the severity of acute experimental colitis, which was characterized by less weight loss, disease activity index, colon shortening, and histology scores, as well as decreased crypt loss and epithelium damage. Furthermore, BHB resulted in significantly increased colonic expression of M2 macrophage-associated genes, including IL-4Ra, IL-10, arginase 1 (Arg-1), and chitinase-like protein 3, following DSS exposure, suggesting an increased M2 macrophage skewing in vivo. Moreover, an in vitro experiment revealed that the addition of BHB directly promoted STAT6 phosphorylation and M2 macrophage-specific gene expression in IL-4-stimulated macrophages. Besides, we found that BHB obviously increased M2 macrophage-induced mucosal repair through promoting intestinal epithelial proliferation. However, the enhancement effect of BHB on M2 macrophage-induced mucosal repair and anti-inflammation was completely inhibited by the STAT6 inhibitor AS1517499. CONCLUSIONS In summary, we show that BHB promotes M2 macrophage polarization through the STAT6-dependent signaling pathway, which contributes to the resolution of intestinal inflammation and the repair of damaged intestinal tissues. Our finding suggests that exogenous BHB supplement may be a useful therapeutic approach for IBD treatment.
Collapse
Affiliation(s)
- Chongyang Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hongbin Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ruo Huang
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinwen Yan
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Mengyao Song
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Gao Tan
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Fachao Zhi
- Guangdong Provincial Key Laboratory of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
159
|
Del Re DP. Hippo-Yap signaling in cardiac and fibrotic remodeling. CURRENT OPINION IN PHYSIOLOGY 2022; 26:100492. [PMID: 36644337 PMCID: PMC9836231 DOI: 10.1016/j.cophys.2022.100492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Cardiac injury initiates a tissue remodeling process in which aberrant fibrosis plays a significant part, contributing to impaired contractility of the myocardium and the progression to heart failure. Fibrotic remodeling is characterized by the activation, proliferation, and differentiation of quiescent fibroblasts to myofibroblasts, and the resulting effects on the extracellular matrix and inflammatory milieu. Molecular mechanisms underlying fibroblast fate decisions and subsequent cardiac fibrosis are complex and remain incompletely understood. Emerging evidence has implicated the Hippo-Yap signaling pathway, originally discovered as a fundamental regulator of organ size, as an important mechanism that modulates fibroblast activity and adverse remodeling in the heart, while also exerting distinct cell type-specific functions that dictate opposing outcomes on heart failure. This brief review will focus on Hippo-Yap signaling in cardiomyocytes, cardiac fibroblasts, and other non-myocytes, and present mechanisms by which it may influence the course of cardiac fibrosis and dysfunction.
Collapse
|
160
|
Hong SM, Lee AY, Hwang SM, Ha YJ, Kim MJ, Min S, Hwang W, Yoon G, Kwon SM, Woo HG, Kim HH, Jeong WI, Shen HM, Im SH, Lee D, Kim YS. NAMPT mitigates colitis severity by supporting redox-sensitive activation of phagocytosis in inflammatory macrophages. Redox Biol 2022; 50:102237. [PMID: 35063804 PMCID: PMC8784331 DOI: 10.1016/j.redox.2022.102237] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/05/2022] [Accepted: 01/12/2022] [Indexed: 12/02/2022] Open
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme in the nicotinamide adenine dinucleotide (NAD+) salvage pathway and plays a crucial role in the maintenance of the NAD+ pool during inflammation. Considering that macrophages are essential for tissue homeostasis and inflammation, we sought to examine the functional impact of NAMPT in inflammatory macrophages, particularly in the context of inflammatory bowel disease (IBD). In this study, we show that mice with NAMPT deletion within the myeloid compartment (Namptf/fLysMCre+/-, Nampt mKO) have more pronounced colitis with lower survival rates, as well as numerous uncleared apoptotic corpses within the mucosal layer. Nampt-deficient macrophages exhibit reduced phagocytic activity due to insufficient NAD+ abundance, which is required to produce NADPH for the oxidative burst. Nicotinamide mononucleotide (NMN) treatment rescues NADPH levels in Nampt mKO macrophages and sustains superoxide generation via NADPH oxidase. Consequently, Nampt mKO mice fail to clear dead cells during tissue repair, leading to substantially prolonged chronic colitis. Moreover, systemic administration of NMN, to supply NAD+, effectively suppresses the disease severity of DSS-induced colitis. Collectively, our findings suggest that activation of the NAMPT-dependent NAD+ biosynthetic pathway, via NMN administration, is a potential therapeutic strategy for managing inflammatory diseases.
Collapse
Affiliation(s)
- Sun Mi Hong
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - A-Yeon Lee
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Sung-Min Hwang
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Yu-Jin Ha
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Moo-Jin Kim
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Seongki Min
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Won Hwang
- MSBIOTECH. LTD, Chungbuk, 27672, Republic of Korea
| | - Gyesoon Yoon
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - So Mee Kwon
- Department of Physiology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Hyun Goo Woo
- Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Physiology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea
| | - Hee-Hoon Kim
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Won-Il Jeong
- Laboratory of Liver Research, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Han-Ming Shen
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, 37673, Republic of Korea; ImmunoBiome, Bio Open Innovation Center, Pohang, 37673, Republic of Korea
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| | - You-Sun Kim
- Department of Biochemistry, Ajou University School of Medicine, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea; Department of Biomedical Sciences, Graduate School of Ajou University, 164 Worldcup-ro, Yeongtong-gu, Suwon, Gyeonggi-do, 16499, Republic of Korea.
| |
Collapse
|
161
|
Ramaccini D, Pedriali G, Perrone M, Bouhamida E, Modesti L, Wieckowski MR, Giorgi C, Pinton P, Morciano G. Some Insights into the Regulation of Cardiac Physiology and Pathology by the Hippo Pathway. Biomedicines 2022; 10:biomedicines10030726. [PMID: 35327528 PMCID: PMC8945338 DOI: 10.3390/biomedicines10030726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 11/16/2022] Open
Abstract
The heart is one of the most fascinating organs in living beings. It beats up to 100,000 times a day throughout the lifespan, without resting. The heart undergoes profound anatomical, biochemical, and functional changes during life, from hypoxemic fetal stages to a completely differentiated four-chambered cardiac muscle. In the middle, many biological events occur after and intersect with each other to regulate development, organ size, and, in some cases, regeneration. Several studies have defined the essential roles of the Hippo pathway in heart physiology through the regulation of apoptosis, autophagy, cell proliferation, and differentiation. This molecular route is composed of multiple components, some of which were recently discovered, and is highly interconnected with multiple known prosurvival pathways. The Hippo cascade is evolutionarily conserved among species, and in addition to its regulatory roles, it is involved in disease by drastically changing the heart phenotype and its function when its components are mutated, absent, or constitutively activated. In this review, we report some insights into the regulation of cardiac physiology and pathology by the Hippo pathway.
Collapse
Affiliation(s)
- Daniela Ramaccini
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Gaia Pedriali
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Mariasole Perrone
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Esmaa Bouhamida
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
| | - Lorenzo Modesti
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Mariusz R. Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland;
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
| | - Paolo Pinton
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| | - Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy; (D.R.); (G.P.); (E.B.)
- Laboratory for Technologies of Advanced Therapies (LTTA), Section of Experimental Medicine, Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (M.P.); (L.M.); (C.G.)
- Correspondence: (P.P.); (G.M.); Tel.: +39-0532-455-802 (P.P.); +39-0532-455-804 (G.M.)
| |
Collapse
|
162
|
Song Y, Dan K, Yao Z, Yang X, Chen B, Hao F. Altered Gut Microbiota in H1-Antihistamine-Resistant Chronic Spontaneous Urticaria Associates With Systemic Inflammation. Front Cell Infect Microbiol 2022; 12:831489. [PMID: 35372130 PMCID: PMC8967245 DOI: 10.3389/fcimb.2022.831489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background and Objective Chronic spontaneous urticaria (CSU) is a histamine-mediated inflammatory skin disease, and second-generation non-sedating H1-antihistamines (nsAH) at licensed doses have long been the first-line therapy in CSU. However, about 50% of patients are resistant to nsAH, and the precise pathogenesis remains largely unknown but seems to be associated with low-level systemic or intestinal inflammation. We aim to determine the fecal microbial composition and clarify its correlation with the clinical profiles og CSU with nsAH resistance. Methods A total of 25 CSU patients with or 19 CSU patients without nsAH resistance and 19 healthy controls (HC) were enrolled in this study. The intestinal microbiome was detected by 16S rRNA sequencing. The data were analyzed using R language software. Results Significantly higher urticarial activity score for 7 days, stool calprotectin, erythrocyte sedimentation rate, serum C-reactive protein, and interleukin-6, but much lower alpha-diversity and evenness of fecal bacterial community were observed in CSU patients with nsAH resistance than in those without (P <0.05 for all variables). Compared to patients with nsAH-responsiveness, the abundance of fecal genera Prevotella, Megamonas, and Escherichia were significantly increased, while that of Blautia, Alistipes, Anaerostipes, and Lachnospira were remarkably reduced in nsAH-resistant patients (uncorrected P <0.05 for all variables). Finally, systemic not intestinal inflammation degree was positively correlated with genera Escherichia, while negatively with genera Blautia, Dorea, Lactobacillus, Eubacterium_hallii_group, and Roseburia. CSU without nsAH resistance and HC individuals showed almost unchanged genera bacterium. Conclusions Among CSU patients, pro-inflammation phenotype relating to enteric dysbacteriosis features nsAH resistance in CSU patients. The results provide clues for future microbial-based or anti-inflammatory therapies on nsAH resistant CSU.
Collapse
Affiliation(s)
- Yao Song
- Department of Pediatrics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kena Dan
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengqiu Yao
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Yang
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bangtao Chen
- Department of Dermatology, Chongqing University Three Gorges Hospital, School of Medicine, Chong University, Chongqing, China
| | - Fei Hao
- Department of Dermatology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
163
|
Bai X, Liu W, Chen H, Zuo T, Wu X. Immune Cell Landscaping Reveals Distinct Immune Signatures of Inflammatory Bowel Disease. Front Immunol 2022; 13:861790. [PMID: 35371090 PMCID: PMC8966033 DOI: 10.3389/fimmu.2022.861790] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Determining how the profile of immune cells varies with their disease subtypes and across lesion locations is critical for understanding the pathogenesis in inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC). To that end, we herein combined the IBD TaMMA framework and the CIBERSORT pipeline to deconvolute the large amount of RNA-seq data from patients with IBD (both CD and UC were included) and healthy human controls across 28 cohorts (a total of 3,852 samples) while accommodating data heterogeneity across cohorts, to define the immune cell landscape of IBD. Our study uncovered that both absolute quantities of innate and adaptive immune cell populations were elevated in most intestinal regions of IBD patients, yet disease-specific (CD versus UC) and intestinal location (ileum, colon, and rectum)-specific features. In the ileum, the increase in innate immune cells was more pronounced in CD than UC. In contrast, innate and adaptive immune cells were elevated more drastically in the UC than CD in the rectum. Such revelation of immune signatures across the highly variable IBD phenotypes (in both disease subtypes and intestinal regions) underpins differential immune-pathophysiological mechanisms in IBD pathogenesis and therefore serves as a resource for the development of future targeted studies.
Collapse
Affiliation(s)
- Xiaowu Bai
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Weixin Liu
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Hongxia Chen
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Zuo
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaojian Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Supported by National Key Clinical Discipline, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
164
|
Liang L, Liu L, Zhou W, Yang C, Mai G, Li H, Chen Y. Gut microbiota-derived butyrate regulates gut mucus barrier repair by activating the macrophage/WNT/ERK signaling pathway. Clin Sci (Lond) 2022; 136:291-307. [PMID: 35194640 DOI: 10.1042/cs20210778] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/10/2022] [Accepted: 01/21/2022] [Indexed: 12/19/2022]
Abstract
Ulcerative colitis (UC) is majorly associated with dysregulation of the dynamic cross-talk among microbial metabolites, intestinal epithelial cells, and macrophages. Several studies have reported the significant role of butyrate in host-microbiota communication. However, whether butyrate provides anti-inflammatory profiles in macrophages, thus contributing to UC intestinal mucus barrier protection, has currently remained elusive. In the current study, we found that butyrate increased mucin production and the proportion of mucin-secreting goblet cells in the colon crypt in a macrophage-dependent manner by using clodronate liposomes. Furthermore, in vivo and in vitro studies were conducted, validating that butyrate facilitates M2 macrophage polarization with the elevated expressions of CD206 and arginase-1 (Arg1). In macrophages/goblet-like LS174T cells co-culture systems, butyrate-primed M2 macrophages significantly enhanced the expression of mucin-2 (MUC2) and SPDEF (goblet cell marker genes) than butyrate alone, while blockade of WNTs secretion or ERK1/2 activation significantly decreased the beneficial effect of butyrate-primed macrophages on goblet cell function. Additionally, the adoptive transfer of butyrate-induced M2 macrophages facilitated the generation of goblet cells and mucus restoration following dextran sulfate sodium (DSS) insult. Taken together, our results revealed a novel mediator of macrophage-goblet cell cross-talk associated with the regulation of epithelial barrier integrity, implying that the microbial metabolite butyrate may serve as a candidate therapeutic target for UC.
Collapse
Affiliation(s)
- Liping Liang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Le Liu
- Department of Gastroenterology, Integrative Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Wanyan Zhou
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chenghai Yang
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Integrative Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Genghui Mai
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haolin Li
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Department of Gastroenterology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Gastroenterology, Integrative Clinical Microecology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
165
|
Zhang Y, Liu Y, Wang J, Jiang Z, Zhang L, Cui Y, Zhao D, Wang Y. Atractylenolide II inhibits tumor-associated macrophages (TAMs)-induced lung cancer cell metastasis. Immunopharmacol Immunotoxicol 2022; 44:227-237. [PMID: 35166628 DOI: 10.1080/08923973.2022.2037629] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVE M2-like tumor-associated macrophages (TAMs) play a crucial role in promoting tumor proliferation, angiogenesis, and metastasis. In the current study, we investigated the relationship between macrophage polarization and the antitumor effect of Atractylenolide II (AT-II) in lung cancer cells. MATERIALS AND METHODS Cell viability, migration, and invasion were determined by MTT assay, wound healing assay, and transwell assay, respectively. Flow cytometry analysis showed the percentage of CD206+ cells. Gene expression was determined by real-time PCR, western blotting, and immunofluorescence staining. Lewis lung carcinoma mouse xenograft and metastasis models were used to examine the effects of AT-II on lung cancer in vivo. RESULTS AT-II (2.5 and 5 µM) did not cause significant inhibition of A549 cell viability but markedly inhibited IL-4/IL-13-induced M2-like polarization, evidenced by the decreased expression of the M2 surface marker CD206, down-regulation of specific M2-marker genes (Arg-1, IL-10 and TGF-β) as well as inhibition of M2 macrophages-mediated invasion and migration of A549 cells. In addition, AT-II inhibited IL-4/IL-13-induced activation of the STAT6 signaling pathway that is vital in the M2-like polarization of macrophages. In animal models, administration of AT-II (50 mg kg-1, i.g., QD for 21 days) significantly inhibited tumor growth, reduced pulmonary metastatic nodules, and down-regulated the percentages of M2 macrophages (F4/80+ and CD206+) in total macrophages (F4/80+) in tumor tissues and pulmonary metastatic nodules. CONCLUSIONS AT-II effectively inhibits M2-like polarization, thereby inhibiting lung cancer cell metastasis both in vivo and in vitro, revealing a novel potential strategy for the antitumor effect of AT-II.
Collapse
Affiliation(s)
- Yunting Zhang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yuxi Liu
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Jianguang Wang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Zongying Jiang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Lin Zhang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yong Cui
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Danyu Zhao
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| | - Yanjie Wang
- College of Integrated Chinese and Western Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, P.R. China
| |
Collapse
|
166
|
Chen R, Pang X, Li L, Zeng Z, Chen M, Zhang S. Ubiquitin-specific proteases in inflammatory bowel disease-related signalling pathway regulation. Cell Death Dis 2022; 13:139. [PMID: 35145062 PMCID: PMC8831562 DOI: 10.1038/s41419-022-04566-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023]
Abstract
The exact pathogenesis of inflammatory bowel disease (IBD), a chronic gastrointestinal inflammatory disease comprising Crohn’s disease and ulcerative colitis, remains unclear. Studies on ubiquitination, which regulates the degradation of inflammation signalling pathway molecules, and deubiquitination have provided novel insights. Targeting the ubiquitin-specific protease (USP) family of deubiquitinases elucidates IBD signalling pathway mechanisms and possibly, IBD therapeutic solutions. Here, we characterised USPs as chief regulators of pro-inflammatory signalling pathways, including nuclear factor-κB and transforming growth factor-β; analysed the relationship between USPs and IBD pathogenesis in terms of genetic susceptibility, intestinal epithelial barrier, immunity, and gut microbiota; and discussed future research prospects.
Collapse
Affiliation(s)
- Rirong Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaobai Pang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Li Li
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhirong Zeng
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shenghong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
167
|
Song H, Yang Y, Sun Y, Wei G, Zheng H, Chen Y, Cai D, Li C, Ma Y, Lin Z, Shi X, Liao W, Liao Y, Zhong L, Bin J. Circular RNA Cdyl promotes abdominal aortic aneurysm formation by inducing M1 macrophage polarization and M1-type inflammation. Mol Ther 2022; 30:915-931. [PMID: 34547461 PMCID: PMC8821928 DOI: 10.1016/j.ymthe.2021.09.017] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/09/2021] [Accepted: 09/14/2021] [Indexed: 02/04/2023] Open
Abstract
Macrophage polarization plays a crucial role in regulating abdominal aortic aneurysm (AAA) formation. Circular RNAs (circRNAs) are important regulators of macrophage polarization during the development of cardiovascular diseases. How-ever, the roles of circRNAs in regulating AAA formation through modulation of macrophage polarization remain unknown. In the present study, we compared circRNA microarray data under two distinct polarizing conditions (M1 and M2 macrophages) and identified an M1-enriched circRNA, circCdyl. Loss- and gain-of-function assay results demonstrated that circCdyl overexpression accelerated angiotensin II (Ang II)- and calcium chloride (CaCl2)-induced AAA formation by promoting M1 polarization and M1-type inflammation, while circCdyl deficiency showed the opposite effects. RNA pulldown, mass spectrometry analysis, and RNA immunoprecipitation (RIP) assays were conducted to elucidate the underlying mechanisms by which circCdyl regulates AAA formation and showed that circCdyl promotes vascular inflammation and M1 polarization by inhibiting interferon regulatory factor 4 (IRF4) entry into the nucleus, significantly inducing AAA formation. In addition, circCdyl was shown to act as a let-7c sponge, promoting C/EBP-δ expression in macrophages to induce M1 polarization. Our results indicate an important role for circCdyl-mediated macrophage polarization in AAA formation and provide a potent therapeutic target for AAA treatment.
Collapse
Affiliation(s)
- Haoyu Song
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Wards of Cadres, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China
| | - Yang Yang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yili Sun
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Guoquan Wei
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Hao Zheng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yijin Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Donghua Cai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Chuling Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Yusheng Ma
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Zhongqiu Lin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Xiaoran Shi
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China
| | - Lintao Zhong
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China,Corresponding author: Lintao Zhong, MD, PhD, Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai 519000, China.
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou 510515, China,Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510005, China,Guangdong Provincial Key Laboratory of Shock and Microcirculation, Guangzhou 510515, China,Corresponding author: Jianping Bin, MD, PhD, Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
168
|
Qing J, Ren Y, Zhang Y, Yan M, Zhang H, Wu D, Ma Y, Chen Y, Huang X, Wu Q, Mazhar M, Wang L, Liu J, Ding BS, Cao Z. Dopamine receptor D2 antagonism normalizes profibrotic macrophage-endothelial crosstalk in non-alcoholic steatohepatitis. J Hepatol 2022; 76:394-406. [PMID: 34648896 DOI: 10.1016/j.jhep.2021.09.032] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 09/16/2021] [Accepted: 09/19/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Currently there is no effective treatment for liver fibrosis, which is one of the main histological determinants of non-alcoholic steatohepatitis (NASH). While Hippo/YAP (Yes-associated protein) signaling is essential for liver regeneration, its aberrant activation frequently leads to fibrosis and tumorigenesis. Unravelling "context-specific" contributions of YAP in liver repair might help selectively bypass fibrosis and preserve the pro-regenerative YAP function in hepatic diseases. METHODS We used murine liver fibrosis and minipig NASH models, and liver biopsies from patients with cirrhosis. Single-cell RNA-sequencing (scRNA-Seq) was performed, and a G-protein-coupled receptor (GPCR) ligand screening system was used to identify cell-selective YAP inhibitors. RESULTS YAP levels in macrophages are increased in the livers of humans and mice with liver fibrosis. The increase in type I interferon and attenuation of hepatic fibrosis observed in mice specifically lacking Yap1 in myeloid cells provided further evidence for the fibrogenic role of macrophage YAP. ScRNA-Seq further showed that defective YAP pathway signaling in macrophages diminished a fibrogenic vascular endothelial cell subset that exhibited profibrotic molecular signatures such as angiocrine CTGF and VCAM1 expression. To specifically target fibrogenic YAP in macrophages, we utilized a GPCR ligand screening system and identified a dopamine receptor D2 (DRD2) antagonist that selectively blocked YAP in macrophages but not hepatocytes. Genetic and pharmacological targeting of macrophage DRD2 attenuated liver fibrosis. In a large animal (minipig) NASH model recapitulating human pathology, the DRD2 antagonist blocked fibrosis and restored hepatic architecture. CONCLUSIONS DRD2 antagonism selectively targets YAP-dependent fibrogenic crosstalk between macrophages and CTGF+VCAM1+ vascular niche, promoting liver regeneration over fibrosis in both rodent and large animal models. LAY SUMMARY Fibrosis in the liver is one of the main histological determinants of non-alcoholic steatohepatitis (NASH), a disease paralleling a worldwide surge in metabolic syndromes. Our study demonstrates that a macrophage-specific deficiency in Yes-associated protein (YAP) attenuates liver fibrosis. Dopamine receptor D2 (DRD2) antagonism selectively blocks YAP in macrophages and thwarts liver fibrosis in both rodent and large animal models, and thus holds potential for the treatment of NASH.
Collapse
Affiliation(s)
- Jie Qing
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China; National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China; Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yafeng Ren
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Yuwei Zhang
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Mengli Yan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Hua Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Dongbo Wu
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, 610064, China
| | - Yongyuan Ma
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Yutian Chen
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Xiaojuan Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Qinkai Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China
| | - Maryam Mazhar
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jian Liu
- Research Center of Integrated Traditional Chinese and Western Medicine, The Affiliated Traditional Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China; Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, New York, 10128, USA.
| | - Zhongwei Cao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610064, China.
| |
Collapse
|
169
|
Xu J, Liu XY, Zhang Q, Liu H, Zhang P, Tian ZB, Zhang CP, Li XY. Crosstalk Among YAP, LncRNA, and Tumor-Associated Macrophages in Tumorigenesis Development. Front Oncol 2022; 11:810893. [PMID: 35071016 PMCID: PMC8770286 DOI: 10.3389/fonc.2021.810893] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022] Open
Abstract
Long non-coding RNAs (ncRNAs), which do not encode proteins, regulate cell proliferation, tumor angiogenesis, and metastasis and are closely associated with the development, progression, and metastasis of many cancers. Tumor-associated macrophages (TAMs) in the tumor microenvironment play an important role in cancer progression. The Hippo signaling pathway regulates cell proliferation and apoptosis, maintains tissue and organ size, and homeostasis of the internal environment of organisms. Abnormal expression of Yes-associated protein (YAP), the Hippo signaling pathway key component, is widely observed in various malignancies. Further, TAM, lncRNA, and YAP are currently valuable targets for cancer immunotherapy. In this review, we have logically summarized recent studies, clarified the close association between the three factors and tumorigenesis, and analyzed the outlook of tumor immunotherapy.
Collapse
Affiliation(s)
- Jing Xu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Innovation Platform of Marine Drug Screening & Evaluation, Qingdao Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xin-Yuan Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hua Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Peng Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zi-Bin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui-Ping Zhang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiao-Yu Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
170
|
Yu H, Hou Z, Chen N, Luo R, Yang L, Miao M, Ma X, Zhou L, He F, Shen Y, Liu X, Wang Y. Yes-associated Protein Contributes to Magnesium Alloy-derived Inflammation in Endothelial Cells. Regen Biomater 2022; 9:rbac002. [PMID: 35480861 PMCID: PMC9039498 DOI: 10.1093/rb/rbac002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 12/25/2021] [Accepted: 01/04/2022] [Indexed: 11/19/2022] Open
Abstract
Magnesium alloy (Mg alloy) has attracted massive attention in the potential applications of cardiovascular stents because of its good biocompatibility and degradability. However, whether and how the Mg alloy induces inflammation in endothelial cells remains unclear. In the present work, we investigated the activation of Yes-associated protein (YAP) upon Mg alloy stimuli and unveiled the transcriptional function in Mg alloy-induced inflammation. Quantitative RT–PCR, western blotting and immunofluorescence staining showed that Mg alloy inhibited the Hippo pathway to facilitate nuclear shuttling and activation of YAP in human coronary artery endothelial cells (HCAECs). Chromatin immunoprecipitation followed sequencing was carried out to explore the transcriptional function of YAP in Mg alloy-derived inflammation. This led to the observation that nuclear YAP further bonded to the promoter region of inflammation transcription factors and co-transcription factors. This binding event activated their transcription and modified mRNA methylation of inflammation-related genes through regulating the expression of N6-methyladenosine modulators (METTL3, METTL14, FTO and WTAP). This then promoted inflammation-related gene expression and aggravated inflammation in HCAECs. In YAP deficiency cells, Mg alloy-induced inflammation was reduced. Collectively, our data suggest that YAP contributes to the Mg alloy-derived inflammation in HCAECs and may provide a potential therapeutic target that alleviates inflammation after Mg alloy stent implantation.
Collapse
Affiliation(s)
- Hongchi Yu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Zhe Hou
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Nuoya Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Rifang Luo
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Li Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| | - Michael Miao
- Division of Oral & Craniofacial Health Sciences, University of North Carolina Adams School of Dentistry, Chapel Hill, NC,27599, USA
| | - Xiaoyi Ma
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Lifeng Zhou
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Fugui He
- Beijing Key Laboratory of Cardiac Drug Device Technology and Evidence Based Medicine, Beijing 100021, China
| | - Yang Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610065, China
| |
Collapse
|
171
|
Jiang S, Zeng Q, Zhao K, Liu J, Sun Q, Huang K, He Y, Zhang X, Wang H, Shi X, Feng C, Deng X, Wei Y. Chirality Bias Tissue Homeostasis by Manipulating Immunological Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105136. [PMID: 34601779 DOI: 10.1002/adma.202105136] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/15/2021] [Indexed: 06/13/2023]
Abstract
The physiological chirality of extracellular environments is substantially affected by pathological diseases. However, how this stereochemical variation drives host immunity remains poorly understood. Here, it is reported that pathology-mimetic M-nanofibrils-but not physiology-mimetic P-nanofibrils-act as a defense mechanism that helps to restore tissue homeostasis by manipulating immunological response. Quantitative multi-omics in vivo and in vitro shows that M-nanofibrils significantly inhibit inflammation and promote tissue regeneration by upregulating M2 macrophage polarization and downstream immune signaling compared with P-nanofibrils. Molecular analysis and theoretical simulation demonstrate that M-chirality displays higher stereo-affinity to cellular binding, which induces higher cellular contractile stress and activates mechanosensitive ion channel PIEZOl to conduct Ca2+ influx. In turn, the nuclear transfer of STAT is biased by Ca2+ influx to promote M2 polarization. These findings underscore the structural mechanisms of disease, providing design basis for immunotherapy with bionic functional materials.
Collapse
Affiliation(s)
- Shengjie Jiang
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Qiang Zeng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kai Zhao
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Jinying Liu
- Key Laboratory for Special Functional Materials of Ministry of Education, School of Materials Science and Engineering, Henan University, Kaifeng, 475004, P. R. China
| | - Qiannan Sun
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Kang Huang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Ying He
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Xuehui Zhang
- Department of Dental Materials and Dental Medical Devices Testing Center, National Engineering Laboratory for Digital and Material Technology of Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
| | - Hui Wang
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Xinghua Shi
- CAS Key Laboratory of Nanosystem and Hierarchical Fabrication, Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, P. R. China
| | - Chuanliang Feng
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiaotong University, Shanghai, 200240, P. R. China
| | - Xuliang Deng
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| | - Yan Wei
- Beijing Laboratory of Biomedical Materials, Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, Beijing, 100081, P. R. China
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, P. R. China
| |
Collapse
|
172
|
Tian G, Liu C, Gong Q, Yu Z, Wang H, Zhang D, Cong H. Human Umbilical Cord Mesenchymal Stem Cells Improve the Necrosis and Osteocyte Apoptosis in Glucocorticoid-Induced Osteonecrosis of the Femoral Head Model through Reducing the Macrophage Polarization. Int J Stem Cells 2021; 15:195-202. [PMID: 34965999 PMCID: PMC9148830 DOI: 10.15283/ijsc21120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/01/2021] [Accepted: 10/25/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives Apoptosis is an outstanding determinant of glucocorticoid (GC)-induced osteonecrosis of the femoral head (ONFH). Human umbilical cord mesenchymal stem cells (hUC-MSCs) have been demonstrated to be associated with apoptosis in diseases models. However, the role of hUC-MSCs in GC-induced ONFH via regulating apoptosis still needs further study. Methods and Results In the present study, a GC-induced ONFH model was built in vivo through a consecutive injection with lipopolysaccharide (LPS) and methylprednisolone. The necrosis and apoptosis of the femoral head was evaluated by histological and Terminal-deoxynucleoitidyl Transferase Mediated Nick End Labeling (TUNEL) assay. The level of collagen and TRAP positive cells were determined by Masson and TRAP staining, respectively. M1 macrophage polarization was assessed using immunofluorescence assay. The level of proinflammatory cytokines including tumor necrosis factor (TNF)‐α, Interleukin (IL)‐1β and IL-6 of femoral head was determined by enzyme-linked immunosorbent assay (ELISA) kits. The protein expression of AKT, mTOR, p-AKT and p-mTOR was detected using western blot assay. The results showed that hUC-MSCs treatment prominently promoted the GC-induced the decrease of the collagen level and the increase of TRAP positive cells. Besides, hUC-MSCs treatment decreased necrosis and apoptosis, macrophage polarization, the level of TNF‐α, IL‐1β and IL-6, the protein expression of p-AKT and p-mTOR, and the radio of p-AKT to AKT and p-mTOR to mTOR of femoral head in vivo. Conclusions Therefore, the present study revealed that hUC-MSCs improved the necrosis and osteocyte apoptosis in GC-induced ONFH model through reducing the macrophage polarization, which was associated with the inhibition of AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Gang Tian
- Department of Orthopedics, Weihai Central Hospital Affiliated to Qingdao University & Qingdao University, Weihai, China
| | - Chuanjie Liu
- Xinxiang Medical University, Xinxiang, China.,Weihai Key Laboratory of Autoimmunity & Central Laboratory of Weihai Central Hospital, Weihai, China
| | - Qi Gong
- Weihai Key Laboratory of Autoimmunity & Central Laboratory of Weihai Central Hospital, Weihai, China
| | - Zhiping Yu
- Department of Sports Medicine, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Haitao Wang
- Department of Trauma Surgery, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Daoqiang Zhang
- Weihai Key Laboratory of Autoimmunity & Central Laboratory of Weihai Central Hospital, Weihai, China
| | - Haibo Cong
- Department of Orthopedics, Weihai Central Hospital Affiliated to Qingdao University & Weihai Key Laboratory of Autoimmunity, Weihai, China
| |
Collapse
|
173
|
Shao X, Sun S, Zhou Y, Wang H, Yu Y, Hu T, Yao Y, Zhou C. Bacteroides fragilis restricts colitis-associated cancer via negative regulation of the NLRP3 axis. Cancer Lett 2021; 523:170-181. [PMID: 34627951 DOI: 10.1016/j.canlet.2021.10.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023]
Abstract
Patients with persistent ulcerative colitis (UC) are at a higher risk of developing colitis-associated cancer (CAC). Previous studies have reported that intestinal microbiota disturbance plays an important role in the process of CAC development in patients with UC, indicating that targeted intervention of intestinal microbiota and its metabolites may be a potential therapeutic strategy. Gut microbiota in the process of colorectal cancer development in UC patients was analyzed using the gutMEGA database and verified in fecal samples. The abundance of Bacteroides fragilis reduced significantly in the process of colitis associated cancer development. Broad-spectrum antibiotics (BSAB) intervene with the intestinal microbiota of mice and accelerate the process of colon cancer development. However, gavage transplantation with B. fragilis can effectively reverse the effects of BSAB. In the intestinal tract, B. fragilis promotes the secretion of short-chain fatty acids (SCFAs). Subsequently, SCFAs, especially butyrate, negatively regulate the inflammatory signaling pathway mediated by NLRP3 to inhibit the activation of macrophages and the secretion of proinflammatory mediators such as IL-18 and IL-1β, reducing the level of intestinal inflammation and restricting CAC development. In conclusion, colonization with B. fragilis has been shown to be effective in ameliorating intestinal epithelial damage caused by chronic inflammation and preventing the development of colonic tumors. Thus, it can be a therapeutic intervention strategy with good clinical application prospects.
Collapse
Affiliation(s)
- Xinyu Shao
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Shishuo Sun
- Cancer Institute, The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuqing Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Huiyu Wang
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yang Yu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Tong Hu
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yizhou Yao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Chunli Zhou
- Department of Gastroenterology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China.
| |
Collapse
|
174
|
Caire R, Audoux E, Courbon G, Michaud E, Petit C, Dalix E, Chafchafi M, Thomas M, Vanden-Bossche A, Navarro L, Linossier MT, Peyroche S, Guignandon A, Vico L, Paul S, Marotte H. YAP/TAZ: Key Players for Rheumatoid Arthritis Severity by Driving Fibroblast Like Synoviocytes Phenotype and Fibro-Inflammatory Response. Front Immunol 2021; 12:791907. [PMID: 34956224 PMCID: PMC8695934 DOI: 10.3389/fimmu.2021.791907] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/18/2021] [Indexed: 12/29/2022] Open
Abstract
Objective The role of YAP/TAZ, two transcriptional co-activators involved in several cancers, was investigated in rheumatoid arthritis (RA). Methods Fibroblast like synoviocytes (FLS) from patients with RA or osteoarthritis were cultured in 2D or into 3D synovial organoids. Arthritis rat model (n=28) and colitis mouse model (n=21) were used. YAP/TAZ transcriptional activity was inhibited by verteporfin (VP). Multiple techniques were used to assess gene and/or protein expression and/or localization, cell phenotype (invasion, proliferation, apoptosis), bone erosion, and synovial stiffness. Results YAP/TAZ were transcriptionally active in arthritis (19-fold increase for CTGF expression, a YAP target gene, in RA vs. OA organoids; p<0.05). Stiff support of culture or pro-inflammatory cytokines further enhanced YAP/TAZ transcriptional activity in RA FLS. Inhibiting YAP/TAZ transcriptional activity with VP restored a common phenotype in RA FLS with a decrease in apoptosis resistance, proliferation, invasion, and inflammatory response. Consequently, VP blunted hyperplasic lining layer formation in RA synovial organoids. In vivo, VP treatment strongly reduced arthritis severity (mean arthritic index at 3.1 in arthritic group vs. 2.0 in VP treated group; p<0.01) by restoring synovial homeostasis and decreasing systemic inflammation. YAP/TAZ transcriptional activity also enhanced synovial membrane stiffening in vivo, thus creating a vicious loop with the maintenance of YAP/TAZ activation over time in FLS. YAP/TAZ inhibition was also effective in another inflammatory model of mouse colitis. Conclusion Our work reveals that YAP/TAZ were critical factors during arthritis. Thus, their transcriptional inhibition could be relevant to treat inflammatory related diseases.
Collapse
Affiliation(s)
- Robin Caire
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Estelle Audoux
- CIRI (Centre International de Recherche en Infectiologie), Equipe GIMAP (Team 15), INSERM, U1111, CNRS, ENS, UCBL1, Université Jean Monnet, Université de Lyon, Saint-Etienne, France
| | | | - Eva Michaud
- CIRI (Centre International de Recherche en Infectiologie), Equipe GIMAP (Team 15), INSERM, U1111, CNRS, ENS, UCBL1, Université Jean Monnet, Université de Lyon, Saint-Etienne, France
| | - Claudie Petit
- INSERM, U1059-SAINBIOSE, Mines Saint-Etienne, Université de Lyon, Saint-Etienne, France
| | - Elisa Dalix
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Marwa Chafchafi
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Mireille Thomas
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | | | - Laurent Navarro
- INSERM, U1059-SAINBIOSE, Mines Saint-Etienne, Université de Lyon, Saint-Etienne, France
| | | | - Sylvie Peyroche
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Alain Guignandon
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Laurence Vico
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France
| | - Stephane Paul
- CIRI (Centre International de Recherche en Infectiologie), Equipe GIMAP (Team 15), INSERM, U1111, CNRS, ENS, UCBL1, Université Jean Monnet, Université de Lyon, Saint-Etienne, France.,CIC INSERM, 1408, Université de Lyon, Saint-Etienne, France
| | - Hubert Marotte
- INSERM, U1059-SAINBIOSE, Université de Lyon, Saint-Etienne, France.,CIC INSERM, 1408, Université de Lyon, Saint-Etienne, France.,Department of Rheumatology, Hôpital Nord, University Hospital Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
175
|
Wang S, Huang J, Liu F, Tan KS, Deng L, Lin Y, Tan W. Isosteviol Sodium Exerts Anti-Colitic Effects on BALB/c Mice with Dextran Sodium Sulfate-Induced Colitis Through Metabolic Reprogramming and Immune Response Modulation. J Inflamm Res 2021; 14:7107-7130. [PMID: 34992409 PMCID: PMC8709797 DOI: 10.2147/jir.s344990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Inflammatory bowel diseases (IBDs) are global health problems that are associated with immune regulation, but clinical IBDs treatment is currently inadequate. Effective preventive or therapeutic methods for immune disorders rely on controlling the function of immune cells. Isosteviol sodium (STV-Na) has antioxidant activity, but the therapeutic effect of STV-Na against IBD remain undocumented. Herein, we investigated the therapeutic effect of STV-Na in mice models with IBDs. METHODS Mice received 3.5% DSS for 7 days to establish IBD models. Intraperitoneal STV-Na was given 2 days before DSS and lasted for 9 days. Commercially available drugs used in treating IBDs (5-aminosalicylic acid, dexamethasone, and infliximab) were used as positive controls. Samples were collected 7 days after colitis induction. Histopathological score, biochemical parameters, molecular biology methods, and metabolomics were used for evaluating the therapeutic effect of STV-Na. RESULTS Our data revealed that STV-Na could significantly alleviate colon inflammation in mice with colitis. Specifically, STV-Na treatment improved body weight loss, increased colon length, decreased histology scores, and restored the hematological parameters of mice with colitis. The untargeted metabolomics analysis revealed that metabolic profiles were restored by STV-Na treatment. Furthermore, STV-Na therapy suppressed the number of CD68 macrophages and F4/80 cell infiltration. And STV-Na suppressed M1 and M2 macrophage numbers along with the mRNA expressions of proinflammatory cytokines. Moreover, STV-Na administration increased the number of regulatory T (Treg) cells while decreasing Th1/Th2/Th17 cell counts in the spleen. Additionally, STV-Na treatment restored intestinal barrier disruption in DSS-triggered colitis tissues by ameliorating the TJ proteins, increasing goblet cell proportions, and mucin protein production, and decreasing the permeability to FITC-dextran, which was accompanied by decreased plasma LPS and DAO contents. CONCLUSION These results indicate that STV-Na can ameliorate colitis by modulating immune responses along with metabolic reprogramming, and could therefore be a promising therapeutic strategy for IBDs.
Collapse
Affiliation(s)
- Shanping Wang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People’s Republic of China
| | - Jiandong Huang
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People’s Republic of China
| | - Fei Liu
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People’s Republic of China
| | - Keai Sinn Tan
- College of Pharmacy, Jinan University, Guangzhou, People’s Republic of China
- Post-Doctoral Innovation Site, Jinan University, Yuanzhi Health Technology Co, Ltd, Zhuhai, People’s Republic of China
| | - Liangjun Deng
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People’s Republic of China
| | - Yue Lin
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, People’s Republic of China
| | - Wen Tan
- Post-Doctoral Innovation Site, Jinan University, Yuanzhi Health Technology Co, Ltd, Zhuhai, People’s Republic of China
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Malaysia
| |
Collapse
|
176
|
Wang L, Dong X, Feng S, Pan H, Jang X, Chen L, Zhao Y, Chen W, Huang Z. VX765 alleviates dextran sulfate sodium-induced colitis in mice by suppressing caspase-1-mediated pyroptosis. Int Immunopharmacol 2021; 102:108405. [PMID: 34865993 DOI: 10.1016/j.intimp.2021.108405] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/15/2021] [Accepted: 11/23/2021] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD) is an autoimmune disease involving intestinal tissue. IBD activates a series of cell death pathways. Pyroptosis is recently identified as a critical cell death pathway in IBD associated with the activation of caspase-1. VX765 is a caspase-1 inhibitor that can be converted to VRT-043198 in vivo. This study was designed to explore the therapeutic effect of VX765 on colitis using a dextran sulfate sodium (DSS)-induced colitis model in mice. In this research, the caspase-1 inhibitor on inflammatory, pyroptosis, apoptosis, macrophage activation, and intestinal barrier were investigated. We found that administration of VX765 attenuated body weight loss, colonic shortening, and colonic pathological injury in mice. Our study also revealed a therapeutic effect of VX765 on colitis in a dose-dependent manner. VX765 inhibited pyroptosis by curbing the Caspase-1/GSDMD pathway and its downstream key inflammatory cytokines--IL-1β and IL-18. These results indicated that VX765 might have a dose-dependent therapeutic effect on DSS-induced colitis in mice.
Collapse
Affiliation(s)
- Li Wang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xubin Dong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Shuyi Feng
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Haoran Pan
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuepei Jang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lifei Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuan Zhao
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Weizhen Chen
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhiming Huang
- Department of Gastroenterology and Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
177
|
Yang D, Zhang N, Li M, Hong T, Meng W, Ouyang T. The Hippo Signaling Pathway: The Trader of Tumor Microenvironment. Front Oncol 2021; 11:772134. [PMID: 34858852 PMCID: PMC8632547 DOI: 10.3389/fonc.2021.772134] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
The Hippo pathway regulates cancer biology in many aspects and the crosstalk with other pathways complicates its role. Accumulated evidence has shown that the bidirectional interactions between tumor cells and tumor microenvironment (TME) are the premises of tumor occurrence, development, and metastasis. The relationship among different components of the TME constitutes a three-dimensional network. We point out the core position of the Hippo pathway in this network and discuss how the regulatory inputs cause the chain reaction of the network. We also discuss the important role of Hippo-TME involvement in cancer treatment.
Collapse
Affiliation(s)
- Duo Yang
- Department of the Forth Clinical Medical College of Nanchang University, Nanchang, China
| | - Na Zhang
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Meihua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Tao Hong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Wei Meng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Taohui Ouyang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
178
|
Fan ZP, Peng ML, Chen YY, Xia YZ, Liu CY, Zhao K, Zhang HP. S100A9 Activates the Immunosuppressive Switch Through the PI3K/Akt Pathway to Maintain the Immune Suppression Function of Testicular Macrophages. Front Immunol 2021; 12:743354. [PMID: 34764959 PMCID: PMC8576360 DOI: 10.3389/fimmu.2021.743354] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 10/12/2021] [Indexed: 01/20/2023] Open
Abstract
Macrophages are functionally plastic and can thus play different roles in various microenvironments. Testis is an immune privileged organ, and testicular macrophages (TMs) show special immunosuppressive phenotype and low response to various inflammatory stimuli. However, the underlying mechanism to maintain the immunosuppressive function of TMs remains unclear. S100A9, a small molecular Ca2+ binding protein, is associated with the immunosuppressive function of macrophages. However, no related research is available about S100A9 in mouse testis. In the present study, we explored the role of S100A9 in TMs. We found that S100A9 was expressed in TMs from postnatal to adulthood and contributed to maintaining the immunosuppressive phenotype of TMs, which is associated with the activation of PI3K/Akt pathway. S100A9 treatment promotes the polarization of bone marrow-derived macrophages from M0 to M2 in vitro. S100A9 was significantly increased in TMs following UPEC-infection and elevated S100A9 contributed to maintain the M2 polarization of TMs. Treatment with S100A9 and PI3K inhibitor decreased the proportion of M2-type TMs in control and UPEC-infected mouse. Our findings reveal a crucial role of S100A9 in maintaining the immunosuppressive function of TMs through the activation of PI3K/Akt pathway, and provide a reference for further understanding the mechanism of immunosuppressive function of TMs.
Collapse
Affiliation(s)
- Zun Pan Fan
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Mei Lin Peng
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Yuan Yao Chen
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Yu Ze Xia
- Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Chun Yan Liu
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | - Hui Ping Zhang
- Institute of Reproductive Health, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| |
Collapse
|
179
|
Liu M, Yan M, He J, Lv H, Chen Z, Peng L, Cai W, Yao F, Chen C, Shi L, Zhang K, Zhang X, Wang DW, Wang L, Zhu Y, Ai D. Macrophage MST1/2 Disruption Impairs Post-Infarction Cardiac Repair via LTB4. Circ Res 2021; 129:909-926. [PMID: 34515499 DOI: 10.1161/circresaha.121.319687] [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] [Indexed: 12/22/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Chemokine CCL2/genetics
- Chemokine CCL2/metabolism
- Chemokine CCL4/genetics
- Chemokine CCL4/metabolism
- Chemokine CXCL2/metabolism
- Female
- Leukotriene B4/metabolism
- Lipoxygenase/metabolism
- Macrophages/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Myocardial Infarction/metabolism
- Myocardium/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Leukotriene B4/antagonists & inhibitors
- Receptors, Leukotriene B4/metabolism
- Serine-Threonine Kinase 3/genetics
- Serine-Threonine Kinase 3/metabolism
Collapse
Affiliation(s)
- Mingming Liu
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital (M.L.)
| | - Meng Yan
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- The First Affiliated Hospital of Soochow University Department of Pathology, Soochow University, Suzhou (M.Y.)
| | - Jinlong He
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Huizhen Lv
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Zhipeng Chen
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Liyuan Peng
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Wenbin Cai
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Fang Yao
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (F.Y., L.W.)
| | - Chen Chen
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Lei Shi
- Biochemistry and Molecular Biology, School of Basic Medical Sciences (L.S., K.Z.), Tianjin Medical University
| | - Kai Zhang
- Biochemistry and Molecular Biology, School of Basic Medical Sciences (L.S., K.Z.), Tianjin Medical University
| | - Xu Zhang
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Dao-Wen Wang
- Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan (L.P., C.C., D.-W.W.)
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College (F.Y., L.W.)
| | - Yi Zhu
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| | - Ding Ai
- Tianjin Key Laboratory of Ion and Molecular Function of Cardiovascular Diseases, Tianjin Institute of Cardiology, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), the Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics (M.L., M.Y., H.L., D.A.), Tianjin Medical University
- Physiology and Pathophysiology (J.H., H.L., Z.C., W.C., X.Z., Y.Z., D.A.), Tianjin Medical University
| |
Collapse
|
180
|
Li Z, Zhang S, Zhang Y, Chen J, Wu F, Liu G, Chen GQ. Applications and Mechanism of 3-Hydroxybutyrate (3HB) for Prevention of Colonic Inflammation and Carcinogenesis as a Food Supplement. Mol Nutr Food Res 2021; 65:e2100533. [PMID: 34704372 DOI: 10.1002/mnfr.202100533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 10/07/2021] [Indexed: 01/19/2023]
Abstract
SCOPE Inflammatory bowel disease and colorectal carcinogenesis (CRC) are common diseases without effective prevention approach. 3-Hydroxybutyrate (3HB) reported to have multiple functions as an oral food supplement. This study observes that 3HB prevents mouse colitis and CRC. METHODS AND RESULTS The sensitivity of wild type (WT) and GPR109a-/- mice to colitis is compared using dextran sulfate sodium salt (DSS)-induced colitis model. Flow cytometry showed that 3HB cellular surface receptor GPR109a that can decrease the percentage of M1 macrophages from 50% of the DSS-induced acute colitis mouse group to 42% DSS+3HB group mediating the inhibitory effect on inflammation. Bone marrow transplantation experiments further demonstrated that the function of 3HB depended on bone marrow cells. Subsequently, the sensitivity of WT and GPR109a-/- mice to CRC is compared using an azoxymethane-DSS-induced CRC mouse model. It is found that the activation of GPR109a inhibited CRC, depended on reduced myeloid-derived suppressor cells accumulation from 27% of the DSS group to 19% of the DSS+3HB group studied using flow cytometry. CONCLUSION It is concluded that 3HB significantly suppresses colonic inflammation and carcinogenesis, promising to benefit colon disease prevention in form of a food supplement.
Collapse
Affiliation(s)
- Zihua Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Shujie Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yudian Zhang
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Jin Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fuqing Wu
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Gang Liu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, 100084, China.,MOE Key Lab of Industrial Biocatalysis, Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
181
|
Periodontal ligament fibroblast-derived exosomes induced by compressive force promote macrophage M1 polarization via Yes-associated protein. Arch Oral Biol 2021; 132:105263. [PMID: 34688132 DOI: 10.1016/j.archoralbio.2021.105263] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/07/2021] [Accepted: 09/09/2021] [Indexed: 01/31/2023]
Abstract
OBJECTIVES This study aimed to investigate the biological roles and mechanisms of compressive force-stimulated periodontal ligament fibroblasts (PDLFs) on polarization of macrophages DESIGN: PDLFs were stimulated with or without static compressive force, and then conditioned medium, high-molecular weight proteins and low-molecular weight proteins were collected to treat THP-1 macrophages. RT-qPCR and flow cytometric analysis were used to evaluate the polarization of macrophages. Exosomes were isolated by ultracentrifugation method and identified via transmission electron microscopy, western-blot and nano-tracking analysis. The protein level of Yes-Associated Protein (YAP) contained in exosomes was detected by western blot. GW4869 and Verteporfin were used to inhibit exosome secretion and YAP- TEA domain transcription factor (TEAD) interaction respectively. RESULTS Exosomes were successfully purified from PDLFs and could be efficiently incorporated into THP-1 macrophages. conditioned medium, HMW proteins and exosomes derived from compressive force-treated PDLFs significantly induce M1 polarization of macrophages. While inhibiting exosomes secretion by GW4869 treatment eliminated the inductive effect. YAP target genes, connective tissue growth factor (CTGF) and cysteine-rich angiogenic inducer 61 (CYR61) were upregulated in macrophages when treated with exosomes derived from compressive force-treated PDLFs (F-Exo). YAP level was elevated in the F-Exo. When macrophages were treated with Verteporfin, expression of YAP target genes and M1 polarization were significantly downregulated. CONCLUSION These results suggested that exosomes derived from compressive force-treated PDLFs promoted the M1 polarization of the THP-1 macrophages. The elevated level of YAP in the exosomes may be a critical factor for this response.
Collapse
|
182
|
Chen J, Cheng J, Zhao C, Zhao B, Mi J, Li W. The Hippo pathway: a renewed insight in the craniofacial diseases and hard tissue remodeling. Int J Biol Sci 2021; 17:4060-4072. [PMID: 34671220 PMCID: PMC8495397 DOI: 10.7150/ijbs.63305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/20/2021] [Indexed: 12/13/2022] Open
Abstract
The Hippo pathway plays an important role in many pathophysiological processes, including cell proliferation and differentiation, cell death, cell migration and invasion. Because of its extensive functions, Hippo pathway is closely related to not only growth and development, but also many diseases, including inflammation and cancer. In this study, the role of Hippo pathway in craniofacial diseases and hard tissue remodeling was reviewed, in attempting to find new research directions.
Collapse
Affiliation(s)
- Jun Chen
- Xiangya School of Stomatology, Central South University, Changsha 410008, China.,Xiangya Stomatological Hospital, Central South University, Changsha 410008, China.,Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Central South University, Changsha 410008, China
| | - Jingyi Cheng
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Cong Zhao
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Boxuan Zhao
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Jia Mi
- Xiangya School of Stomatology, Central South University, Changsha 410008, China
| | - Wenjie Li
- Xiangya School of Stomatology, Central South University, Changsha 410008, China.,Xiangya Stomatological Hospital, Central South University, Changsha 410008, China.,Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Central South University, Changsha 410008, China.,National Key Laboratory of Science and Technology on High-strength Structural Materials, Central South University, Changsha 410083, China.,State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| |
Collapse
|
183
|
Zhao C, Qiu P, Li M, Liang K, Tang Z, Chen P, Zhang J, Fan S, Lin X. The spatial form periosteal-bone complex promotes bone regeneration by coordinating macrophage polarization and osteogenic-angiogenic events. Mater Today Bio 2021; 12:100142. [PMID: 34647005 PMCID: PMC8495177 DOI: 10.1016/j.mtbio.2021.100142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Bone defects associated with soft tissue injuries are an important cause of deformity that threatens people’s health and quality of life. Although bone substitutes have been extensively explored, effective biomaterials that can coordinate early inflammation regulation and subsequent repair events are still lacking. We prepared a spatial form periosteal bone extracellular matrix (ECM) scaffold, which has advantages in terms of low immunogenicity, good retention of bioactive ingredients, and a natural spatial structure. The periosteal bone ECM scaffold with the relatively low-stiffness periosteum (41.6 ± 3.7 kPa) could inhibit iNOS and IL-1β expression, which might be related to actin-mediated YAP translocation. It also helped to promote CD206 expression with the potential influence of proteins related to immune regulation. Moreover, the scaffold combined the excellent properties of decalcified bone and periosteum, promoted the formation of blood vessels, and good osteogenic differentiation (RUNX2, Col 1α1, ALP, OPN, and OCN), and achieved good repair of a cranial defect in rats. This scaffold, with its natural structural and biological advantages, provides a new idea for bone healing treatment that is aligned with bone physiology. We provided a spatial form periosteal-bone complex. The scaffold preserved major biological components and spatial structure. The periosteum part of the scaffold acted as a physical barrier. The scaffold participated in the transformation of the macrophage phenotype. The scaffold promoted osteogenesis and angiogenesis.
Collapse
Affiliation(s)
- C. Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - P. Qiu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - M. Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - K. Liang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - Z. Tang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - P. Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - J. Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
| | - S. Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Corresponding author.
| | - X. Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, China
- Corresponding author.
| |
Collapse
|
184
|
Zhou Y, Chen S, Gu W, Sun X, Wang L, Tang L. Sinomenine hydrochloride ameliorates dextran sulfate sodium-induced colitis in mice by modulating the gut microbiota composition whilst suppressing the activation of the NLRP3 inflammasome. Exp Ther Med 2021; 22:1287. [PMID: 34630642 PMCID: PMC8461516 DOI: 10.3892/etm.2021.10722] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/02/2021] [Indexed: 12/13/2022] Open
Abstract
Sinomenine is a pure alkaloid that can be isolated from the root of Sinomenium acutum and has been found to exert anti-inflammatory and immunosuppressive effects. The present study investigated the effects of sinomenine hydrochloride (SIN) on inflammation and the gut microbiota composition in the colon of mouse models of dextran sulfate sodium (DSS)-induced colitis. DSS-induced mice colitis was established by treating the mice with drinking water containing 3% (w/v) DSS for 7 days. The disease activity index of each mouse was calculated on a daily basis. All mice were sacrificed on day 11, then the weight of their spleen and length of their colons were measured. The histological analysis was measured by hematoxylin-eosin staining. Oral administration of SIN (100 mg/kg/day) attenuated the DSS-induced increases in the disease activity indices and spleen indices, DSS-induced shortening of the colon length and histological damage. In addition, reverse transcription-quantitative PCR data showed that SIN treatment effectively regulated the expression of inflammatory mediators, specifically by suppressing the expression of proinflammatory gene (TNF-α, IL-6 and inducible nitric oxide synthase) whilst increasing those associated with inhibiting inflammation (IL-10 and arginine 1). Gut microbiota analysis was conducted using 16S ribosomal DNA sequencing. The results revealed that SIN improved bacterial community homeostasis and diversity, which were damaged by DSS. Furthermore, western blotting showed that the activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome was markedly suppressed by SIN treatment. In conclusion, these results indicated that SIN may ameliorate experimental colitis by modulating the gut microbiota composition and suppressing the activation of the NLRP3 inflammasome in mice. Overall, these findings suggested a broad protective effect of SIN in treating inflammatory gut diseases, including ulcerative colitis.
Collapse
Affiliation(s)
- Yan Zhou
- Central Laboratory, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China.,Department of Gastrointestinal Surgery, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Shuai Chen
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Wenxian Gu
- Department of Pathology, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Xiao Sun
- Central Laboratory, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Linxiao Wang
- Central Laboratory, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| | - Liming Tang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Nanjing Medical University, Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
185
|
Li Y, Jiang MY, Chen JY, Xu ZW, Zhang JW, Li T, Zhang LL, Wei W. CP-25 exerts therapeutic effects in mice with dextran sodium sulfate-induced colitis by inhibiting GRK2 translocation to downregulate the TLR4-NF-κB-NLRP3 inflammasome signaling pathway in macrophages. IUBMB Life 2021; 73:1406-1422. [PMID: 34590407 DOI: 10.1002/iub.2564] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022]
Abstract
Deficiency of G protein-coupled receptor kinase 2 (GRK2) was found to protect mice from dextran sulfate sodium (DSS)-induced colitis. Paeoniflorin-6'-O-benzene sulfonate (CP-25) has been shown to exert anti-inflammatory immune regulatory effects in animal models of inflammatory autoimmune disease. This study aimed to investigate the of GRK2 in the pathogenesis of ulcerative colitis (UC) and its effects on macrophage polarization, macrophage subtype regulation of intestinal barrier function, and therapeutic effects of CP-25 in mice with DSS-induced colitis. We found imbalanced macrophage polarization, intestinal barrier dysfunction, and abnormal activation of GRK2 and TLR4-NF-κB-NLRP3 inflammasome signaling pathway in the colonic mucosa of patients with UC. CP-25, restored the damaged intestinal barrier function by inhibiting the transmembrane region of GRK2 in macrophages stimulated by lipopolysaccharides. CP-25 exerted therapeutic effects by ameliorating clinical manifestation, regulating macrophage polarization, and restoring abnormally activated TLR4-NF-κB-NLRP3 inflammasome signaling pathway by inhibiting GRK2. These data suggest the pathogenesis of UC may be related to the imbalance of macrophage polarization, which leads to abnormal activation of TLR4-NF-κB-NLRP3 inflammasome signaling pathway mediated by GRK2 and destruction of the intestinal mucosal barrier. CP-25 confers therapeutic effects on colitis by inhibiting GRK2 translocation to induce the downregulation of TLR4-NF-κB-NLRP3 inflammasome signaling in macrophages.
Collapse
Affiliation(s)
- Ying Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China
| | - Meng-Ya Jiang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China
| | - Jing-Yu Chen
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China
| | - Zhou-Wei Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jia-Wei Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China.,Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tao Li
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China.,Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling-Ling Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Collaborative Innovation Center of Anti-inflammatory and Immune Medicines, Hefei, China
| |
Collapse
|
186
|
Shim J, Goldsmith KC. A New Player in Neuroblastoma: YAP and Its Role in the Neuroblastoma Microenvironment. Cancers (Basel) 2021; 13:cancers13184650. [PMID: 34572875 PMCID: PMC8472533 DOI: 10.3390/cancers13184650] [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: 08/18/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroblastoma is the most common extra-cranial pediatric solid tumor that accounts for more than 15% of childhood cancer-related deaths. High risk neuroblastomas that recur during or after intense multimodal therapy have a <5% chance at a second sustained remission or cure. The solid tumor microenvironment (TME) has been increasingly recognized to play a critical role in cancer progression and resistance to therapy, including in neuroblastoma. The Yes-Associated Protein (YAP) in the Hippo pathway can regulate cancer proliferation, tumor initiation, and therapy response in many cancer types and as such, its role in the TME has gained interest. In this review, we focus on YAP and its role in neuroblastoma and further describe its demonstrated and potential effects on the neuroblastoma TME. We also discuss the therapeutic strategies for inhibiting YAP in neuroblastoma.
Collapse
Affiliation(s)
- Jenny Shim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
| | - Kelly C. Goldsmith
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA;
- Aflac Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
- Correspondence: ; Tel.: +1-404-727-2655
| |
Collapse
|
187
|
Sun C, He B, Sun M, Lv X, Wang F, Chen J, Zhang J, Ye Z, Wen J, Liu P. Yes-Associated Protein in Atherosclerosis and Related Complications: A Potential Therapeutic Target That Requires Further Exploration. Front Cardiovasc Med 2021; 8:704208. [PMID: 34513949 PMCID: PMC8430249 DOI: 10.3389/fcvm.2021.704208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
Atherosclerosis and its complications diseases remain leading causes of cardiovascular morbidity and mortality, bringing a massive burden on public health worldwide. Atherosclerosis is recognized as chronic inflammation, and involves several highly correlated processes, including lipid metabolism dysfunction, endothelial cell dysfunction, inflammation, oxidative stress, vascular smooth muscle cell activation, platelet activation, thrombosis, altered matrix metabolism, and vascular remodeling. Within the past few decades, accumulating evidence has shown that the Yes-associated protein (YAP), the major effector of the Hippo pathway, can play a crucial role in pathogenesis and development of atherosclerosis. Activation of YAP-related pathways, which are induced by alerting flow pattern and matrix stiffness among others, can regulate processes including vascular endothelial cell dysfunction, monocyte infiltration, and smooth muscle cell migration, which contribute to atherosclerotic lesion formation. Further, YAP potentially modulates atherosclerotic complications such as vascular calcification and intraplaque hemorrhage, which require further investigation. Here, we summarized the relevant literature to outline current findings detailing the relationship between of YAP and atherosclerosis and highlight areas for future research.
Collapse
Affiliation(s)
- Congrui Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Bin He
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Mingsheng Sun
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Feng Wang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jie Chen
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianbin Zhang
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhidong Ye
- Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jianyan Wen
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Peng Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.,Department of Cardiovascular Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
188
|
Yang Z, Zhang J, Wang Y, Lu J, Sun Q. Caveolin-1 Deficiency Protects Mice Against Carbon Tetrachloride-Induced Acute Liver Injury Through Regulating Polarization of Hepatic Macrophages. Front Immunol 2021; 12:713808. [PMID: 34434195 PMCID: PMC8380772 DOI: 10.3389/fimmu.2021.713808] [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: 05/24/2021] [Accepted: 07/26/2021] [Indexed: 01/01/2023] Open
Abstract
Polarization of hepatic macrophages plays a crucial role in the injury and repair processes of acute and chronic liver diseases. However, the underlying molecular mechanisms remain elusive. Caveolin-1 (Cav1) is the structural protein of caveolae, the invaginations of the plasma membrane. It has distinct functions in regulating hepatitis, cirrhosis, and hepatocarcinogenesis. Given the increasing number of cases of liver cancer, nonalcoholic steatohepatitis, and non-alcoholic fatty liver disease worldwide, investigations on the role of Cav1 in liver diseases are warranted. In this study, we aimed to investigate the role of Cav1 in the pathogenesis of acute liver injury. Wild-type (WT) and Cav1 knockout (KO) mice (Cav1tm1Mls) were injected with carbon tetrachloride (CCl4). Cav1 KO mice showed significantly reduced degeneration, necrosis, and apoptosis of hepatocytes and decreased level of alanine transaminase (ALT) compared to WT mice. Moreover, Cav1 was required for the recruitment of hepatic macrophages. The analysis of the mRNA levels of CD86, tumor necrosis factor (TNF), and interleukin (IL)-6, as well as the protein expression of inducible nitric oxide synthase (iNOS), indicated that Cav1 deficiency inhibited the polarization of hepatic macrophages towards the M1 phenotype in the injured liver. Consistent with in vivo results, the expressions of CD86, TNF, IL-6, and iNOS were significantly downregulated in Cav1 KO macrophages. Also, fluorescence-activated cell sorting (FACS) analysis showed that the proportion of M1 macrophages was significantly decreased in the liver tissues obtained from Cav1 KO mice following CCl4 treatment. In summary, our results showed that Cav1 deficiency protected mice against CCl4-induced acute liver injury by regulating polarization of hepatic macrophages. We provided direct genetic evidence that Cav1 expressed in hepatic macrophages contributed to the pathogenesis of acute liver injury by regulating the polarization of hepatic macrophages towards the M1 phenotype. These findings suggest that Cav1 expressed in macrophages may represent a potential therapeutic target for acute liver injury.
Collapse
Affiliation(s)
- Ziheng Yang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jie Zhang
- Laboratory Animal Center, Capital Medical University, Beijing, China
| | - Yan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jing Lu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Laboratory Animal Center, Capital Medical University, Beijing, China
| | - Quan Sun
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Laboratory Animal Center, Capital Medical University, Beijing, China
| |
Collapse
|
189
|
Chen Y, Lei J, He S. m 6A Modification Mediates Mucosal Immune Microenvironment and Therapeutic Response in Inflammatory Bowel Disease. Front Cell Dev Biol 2021; 9:692160. [PMID: 34422815 PMCID: PMC8378837 DOI: 10.3389/fcell.2021.692160] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Accumulating evidence links m6A modification with immune infiltration. However, the correlation and mechanism by which m6A modification promotes intestinal immune infiltration in inflammatory bowel disease (IBD) is unknown. Here, genomic information from IBD tissues was integrated to evaluate disease-related m6A modification, and the correlation between the m6A modification pattern and the immune microenvironment in the intestinal mucosa was explored. Next, we identified hub genes from the key modules of the m6Acluster and analyzed the correlation among the hub genes, immune infiltration, and therapy. We found that IGF2BP1 and IGF2BP2 expression was decreased in Crohn's disease (CD) tissues and that IGF2BP2 was decreased in ulcerative colitis (UC) tissues compared with normal tissues (P < 0.05). m6Acluster2, containing higher expressions of IL15, IL16, and IL18, was enriched in M0 macrophage, M1 macrophage, native B cells, memory B cells, and m6Acluster1 with high expression of IL8 and was enriched in resting dendritic and plasma cells (P < 0.05). Furthermore, we reveal that expression of m6A phenotype-related hub genes (i.e., NUP37, SNRPG, H2AFZ) was increased with a high abundance of M1 macrophages, M0 macrophages, and naive B cells in IBD (P < 0.01). Immune checkpoint expression in the genecluster1 with higher expression of hub genes was increased. The anti-TNF therapeutic response of patients in genecluster1 was more significant, and the therapeutic effect of CD was better than that of UC. These findings indicate that m6A modification may affect immune infiltration and therapeutic response in IBD. Assessing the expression of m6A phenotype-related hub genes might guide the choice of IBD drugs and improve the prediction of therapeutic response to anti-TNF therapy.
Collapse
Affiliation(s)
- Yongyu Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Lei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song He
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
190
|
Park BO, Kim SH, Kim JH, Kim SY, Park BC, Han SB, Park SG, Kim JH, Kim S. The Short-Chain Fatty Acid Receptor GPR43 Modulates YAP/TAZ via RhoA. Mol Cells 2021; 44:458-467. [PMID: 34112743 PMCID: PMC8334349 DOI: 10.14348/molcells.2021.0021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/30/2021] [Accepted: 05/27/2021] [Indexed: 01/21/2023] Open
Abstract
GPR43 (also known as FFAR2 or FFA2) is a G-protein-coupled receptor primarily expressed in immune cells, enteroendocrine cells and adipocytes that recognizes short-chain fatty acids, such as acetate, propionate, and butyrate, likely to be implicated in innate immunity and host energy homeostasis. Activated GPR43 suppresses the cAMP level and induces Ca2+ flux via coupling to Gαi and Gαq families, respectively. Additionally, GPR43 is reported to facilitate phosphorylation of ERK through G-protein-dependent pathways and interacts with β-arrestin 2 to inhibit NF-κB signaling. However, other G-protein-dependent and independent signaling pathways involving GPR43 remain to be established. Here, we have demonstrated that GPR43 augments Rho GTPase signaling. Acetate and a synthetic agonist effectively activated RhoA and stabilized YAP/TAZ transcriptional coactivators through interactions of GPR43 with Gαq/11 and Gα12/13. Acetate-induced nuclear accumulation of YAP was blocked by a GPR43-specific inverse agonist. The target genes induced by YAP/TAZ were further regulated by GPR43. Moreover, in THP-1-derived M1-like macrophage cells, the Rho-YAP/TAZ pathway was activated by acetate and a synthetic agonist. Our collective findings suggest that GPR43 acts as a mediator of the Rho-YAP/TAZ pathway.
Collapse
Affiliation(s)
- Bi-Oh Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Seong Heon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biomolecular Science, KRIBB School of Biological Science, UST, Daejeon 34113, Korea
| | - Jong Hwan Kim
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon 34141, Korea
| | - Seon-Young Kim
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Byoung Chul Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Proteome Structural Biology, KRIBB School of Biological Science, UST, Daejeon 34113, Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28160, Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Jeong-Hoon Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Functional Genomics, KRIBB School of Biological Science, Korea University of Science and Technology (UST), Daejeon 34113, Korea
| | - Sunhong Kim
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea
- Department of Biomolecular Science, KRIBB School of Biological Science, UST, Daejeon 34113, Korea
- Present address: Drug Discovery Center, Life Sciences, LG Chem., Seoul 07796, Korea
| |
Collapse
|
191
|
Wimmers F, Donato M, Kuo A, Ashuach T, Gupta S, Li C, Dvorak M, Foecke MH, Chang SE, Hagan T, De Jong SE, Maecker HT, van der Most R, Cheung P, Cortese M, Bosinger SE, Davis M, Rouphael N, Subramaniam S, Yosef N, Utz PJ, Khatri P, Pulendran B. The single-cell epigenomic and transcriptional landscape of immunity to influenza vaccination. Cell 2021; 184:3915-3935.e21. [PMID: 34174187 PMCID: PMC8316438 DOI: 10.1016/j.cell.2021.05.039] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/15/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022]
Abstract
Emerging evidence indicates a fundamental role for the epigenome in immunity. Here, we mapped the epigenomic and transcriptional landscape of immunity to influenza vaccination in humans at the single-cell level. Vaccination against seasonal influenza induced persistently diminished H3K27ac in monocytes and myeloid dendritic cells (mDCs), which was associated with impaired cytokine responses to Toll-like receptor stimulation. Single-cell ATAC-seq analysis revealed an epigenomically distinct subcluster of monocytes with reduced chromatin accessibility at AP-1-targeted loci after vaccination. Similar effects were observed in response to vaccination with the AS03-adjuvanted H5N1 pandemic influenza vaccine. However, this vaccine also stimulated persistently increased chromatin accessibility at interferon response factor (IRF) loci in monocytes and mDCs. This was associated with elevated expression of antiviral genes and heightened resistance to the unrelated Zika and Dengue viruses. These results demonstrate that vaccination stimulates persistent epigenomic remodeling of the innate immune system and reveal AS03's potential as an epigenetic adjuvant.
Collapse
Affiliation(s)
- Florian Wimmers
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michele Donato
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alex Kuo
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tal Ashuach
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shakti Gupta
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093, USA
| | - Chunfeng Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mai Dvorak
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mariko Hinton Foecke
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sarah E Chang
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Thomas Hagan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sanne E De Jong
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Peggie Cheung
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mario Cortese
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven E Bosinger
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Mark Davis
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA 30030, USA
| | - Shankar Subramaniam
- Department of Bioengineering, University of California, San Diego, 9500 Gilman Drive MC 0412, La Jolla, CA 92093, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Chan-Zuckerberg Biohub, San Francisco, CA, USA
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Division of Biomedical Informatics Research, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
192
|
Gianluca C. From good to bad fibroblasts: New promising targets to cure Crohn's disease. EBioMedicine 2021; 70:103483. [PMID: 34280782 PMCID: PMC8318995 DOI: 10.1016/j.ebiom.2021.103483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 06/25/2021] [Indexed: 11/24/2022] Open
Affiliation(s)
- Cipriani Gianluca
- Mayo Clinic, Department of Medicine, Division of Gastroenterology and Hepatology, Enteric Neuroscience Program.
| |
Collapse
|
193
|
He X, Tan S, Shao Z, Wang X. Latitudinal and longitudinal regulation of tissue macrophages in inflammatory diseases. Genes Dis 2021; 9:1194-1207. [PMID: 35873033 PMCID: PMC9293718 DOI: 10.1016/j.gendis.2021.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 01/09/2023] Open
Abstract
Macrophages are dominant innate immune cells. They demonstrate remarkable heterogeneity and plasticity that are essential for homeostasis and host defense. The heterogeneity of tissue macrophages is shaped by the ontogeny, tissue factors, and environmental signals, a pattern in a tissue-associated latitudinal manner. At the same time, macrophages have long been considered as mainly plastic cells. These cells respond to stimulation quickly and in a stimulus-specific way by utilizing a longitudinal cascaded activation, including coordination of signal transducer, epigenetic elements, and transcription factors, conclusively determine the macrophage phenotypes and functions. With the development of cutting-edge technologies, such as fate-mapping, single-cell transcriptomics, ipsc platform, nanotherapeutic materials, etc., our understanding of macrophage biology and the roles in the pathogenesis of diseases is much advanced. This review summarizes recent progress on the latitudinal and longitudinal regulation of tissue macrophages in inflammatory diseases. The latitudinal regulation covers the tissue macrophage origins, tissue factors, and environmental signals, reflecting the macrophage heterogeneity. The longitudinal regulation focuses on how multiple factors shape the phenotypes and functions of macrophage subsets to gain plasticity in inflammatory diseases (i.e., inflammatory bowel disease). In addition, how to target macrophages as a potential therapeutic approach and cutting edge-technologies for tissue macrophage study are also discussed in this review.
Collapse
Affiliation(s)
- XiaoYi He
- Department of Neurology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, PR China
| | - Stephanie Tan
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zhong Shao
- The Third Hospital of Fushun, Fushun, Liaoning 113004, PR China
| | - Xiao Wang
- Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA,Corresponding author. Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago 225 E. Chicago Avenue, Chicago, IL 60611, USA. Fax: +(312) 503 7177.
| |
Collapse
|
194
|
Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease. Inflamm Res 2021; 70:753-764. [PMID: 34212215 DOI: 10.1007/s00011-021-01482-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 01/05/2023] Open
Abstract
Janus kinase/signal transduction and transcriptional activator (JAK/STAT) signaling pathway is a transport hub for cytokine secretion and exerts its effects. The activation of JAK/STAT signaling pathway is essential for the regulation of inflammatory responses. Inappropriate activation or deletion of JAK/STAT signaling pathway is the initiator of the inflammatory response. JAK/STAT signaling pathway has been demonstrated to be involved in the process of innate and adaptive immune response to inflammatory bowel disease (IBD). In this review, we discuss the role of the JAK/STAT signaling pathway in the regulation of different cells in IBD, as well as new findings on the involvement of the JAK/STAT signaling pathway in the regulation of the intestinal immune response. The current status of JAK inhibitors in the treatment of IBD is summarized as well. This review highlights natural remedies that can serve as potential JAK inhibitors. These phytochemicals may be useful in the identification of precursor compounds in the process of designing and developing novel JAK inhibitors.
Collapse
|
195
|
Zheng W, Zhang S, Jiang S, Huang Z, Chen X, Guo H, Li M, Zheng S. Evaluation of immune status in testis and macrophage polarization associated with testicular damage in patients with nonobstructive azoospermia. Am J Reprod Immunol 2021; 86:e13481. [PMID: 34192390 DOI: 10.1111/aji.13481] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/08/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Immune cells residing in the testicular interstitial space form the immunological microenvironment of the testis. They are assumed to play a role in maintaining testicular homeostasis and immune privilege. However, the immune status and related cell polarization in patients with nonobstructive azoospermia (NOA) remains poorly characterized. System evaluation of the testis immunological microenvironment in NOA patients may help to reveal the mechanisms of idiopathic azoospermia. STUDY DESIGN The gene expression patterns of immune cells in normal human testes were systematically analyzed by single-cell RNA sequencing (scRNA-seq) and preliminarily verification by the human protein atlas (HPA) online database. The immune cell infiltration profiles and immune status of patients with NOA was analyzed by single-sample gene set enrichment analysis (ssGSEA) and gene set variation analysis (GSVA) based on four independent public microarray datasets (GSE45885, GSE45887, GSE9210, and GSE145467), obtained from Gene Expression Omnibus (GEO) online database. The relationship between immune cells and spermatogenesis score was further analyzed by Spearman correlation analysis. Finally, immunohistochemistry (IHC) staining was performed to identify the main immune cell types and their polarization status in patients with NOA. RESULTS Both scRNA-seq and HPA analysis showed that testicular macrophages represent the largest pool of immune cells in the normal testis, and also exhibit an attenuated inflammatory response by expressing high levels of tolerance proteins (CD163, IL-10, TGF-β, and VEGF) and reduced expression of TLR signaling pathway-related genes. Correlation analysis revealed that the testicular immune score and macrophages including M1 and M2 macrophages were significantly negatively correlated with spermatogenesis score in patients with NOA (GSE45885 and GSE45887). In addition, the number of M1 and M2 macrophages was significantly higher in patients with NOA (GSE9210 and GSE145467) than in normal testis. GSVA analysis indicated that the immunological microenvironment in NOA tissues was manifested by activated immune system and pro-inflammatory status. IHC staining results showed that the number of M1 and M2 macrophages was significantly higher in NOA tissues than in normal testis and negatively correlated with the Johnson score. CONCLUSION Testicular macrophage polarization may play a vital role in NOA development and is a promising potential therapeutic target.
Collapse
Affiliation(s)
- Wenzhong Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Shaoqin Jiang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhangcheng Huang
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaobao Chen
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Huan Guo
- Department of Urology, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Mengqiang Li
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Song Zheng
- Department of Urology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
196
|
Morciano G, Vezzani B, Missiroli S, Boncompagni C, Pinton P, Giorgi C. An Updated Understanding of the Role of YAP in Driving Oncogenic Responses. Cancers (Basel) 2021; 13:cancers13123100. [PMID: 34205830 PMCID: PMC8234554 DOI: 10.3390/cancers13123100] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary In 2020, the global cancer database GLOBOCAN estimated 19.3 million new cancer cases worldwide. The discovery of targeted therapies may help prognosis and outcome of the patients affected, but the understanding of the plethora of highly interconnected pathways that modulate cell transformation, proliferation, invasion, migration and survival remains an ambitious goal. Here we propose an updated state of the art of YAP as the key protein driving oncogenic response via promoting all those steps at multiple levels. Of interest, the role of YAP in immunosuppression is a field of evolving research and growing interest and this summary about the current pharmacological therapies impacting YAP serves as starting point for future studies. Abstract Yes-associated protein (YAP) has emerged as a key component in cancer signaling and is considered a potent oncogene. As such, nuclear YAP participates in complex and only partially understood molecular cascades that are responsible for the oncogenic response by regulating multiple processes, including cell transformation, tumor growth, migration, and metastasis, and by acting as an important mediator of immune and cancer cell interactions. YAP is finely regulated at multiple levels, and its localization in cells in terms of cytoplasm–nucleus shuttling (and vice versa) sheds light on interesting novel anticancer treatment opportunities and putative unconventional functions of the protein when retained in the cytosol. This review aims to summarize and present the state of the art knowledge about the role of YAP in cancer signaling, first focusing on how YAP differs from WW domain-containing transcription regulator 1 (WWTR1, also named as TAZ) and which upstream factors regulate it; then, this review focuses on the role of YAP in different cancer stages and in the crosstalk between immune and cancer cells as well as growing translational strategies derived from its inhibitory and synergistic effects with existing chemo-, immuno- and radiotherapies.
Collapse
|
197
|
Du XX, He C, Lu X, Guo YL, Chen ZH, Cai LJ. YAP/STAT3 promotes the immune escape of larynx carcinoma by activating VEGFR1-TGFβ signaling to facilitate PD-L1 expression in M2-like TAMs. Exp Cell Res 2021; 405:112655. [PMID: 34044017 DOI: 10.1016/j.yexcr.2021.112655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/20/2021] [Accepted: 04/27/2021] [Indexed: 12/30/2022]
Abstract
Larynx carcinoma (LC) is the most prevalent head and neck cancer among adults. LC xenograft mouse model was generated to verify the effect of VEGF on macrophage polarization and tumor growth in vivo. EdU assay was performed to measure the cell proliferation. Transwell assay was applied to assess cell migration. The expression of YAP and STAT3 was also significantly increased in LC tumor tissues. Moreover, both YAP and STAT3 overexpression in LC cells promoted the proliferation, migration, as well as the secretion of PD-L1 in M2-like TAMs. Mechanistically, the interaction between YAP and STAT3 facilitated the transcription of VEGF. Moreover, with a co-culture system, VEGF secretion in LC cells enhanced PD-L1 expression in M2-like TAMs via activating VEGFR1-TGFβ signaling pathway. Furthermore, VEGF secreted from LC cells also promoted the tumor growth of LC in vivo. We revealed that dysregulated YAP/STAT3 activity in LC cells could enhance the secretion of VEGF, which then functioned on M2-like TAMs via activating VEGFR1-TGFββ pathway to promote the expression of PD-L1 and immunosuppressive function of M2-like TAMs. Therefore, VEGF and PD-L1 might have a pivotal crosstalk between M2-like TAMs and LC cells, which provided a novel therapeutic target in regulating the metastasis of LC in future.
Collapse
Affiliation(s)
- Xiao-Xiao Du
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Key Laboratory of Organ Transplantation, Ministry of Education & NHC Key Laboratory of Organ Transplantation & Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, 430030, PR China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China & Henan Key Laboratory of Digestive Organ Transplantation & Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities & ZhengZhou Key Laboratory of Hepatobiliary, Zhengzhou, 450052, PR China
| | - Chao He
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiang Lu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yu-Liang Guo
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Key Laboratory of Organ Transplantation, Ministry of Education & NHC Key Laboratory of Organ Transplantation & Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, 430030, PR China
| | - Zhong-Hua Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Key Laboratory of Organ Transplantation, Ministry of Education & NHC Key Laboratory of Organ Transplantation & Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, 430030, PR China
| | - Lan-Jun Cai
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
198
|
Anterior Gradient Protein 2 Promotes Mucosal Repair in Pediatric Ulcerative Colitis. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6483860. [PMID: 34055987 PMCID: PMC8149229 DOI: 10.1155/2021/6483860] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 05/05/2021] [Indexed: 12/14/2022]
Abstract
Mucosal healing comprises a key goal of ulcerative colitis (UC) treatment. Anterior gradient protein 2 (AGR2) plays an important role in maintaining intestinal homeostasis in UC. However, the role of AGR2 in the repair of mucosal injury is not yet clear. This study is aimed at investigating the expression of AGR2 in the intestinal tissues of children with UC and its role in repairing mucosal injury. Forty UC patients who were hospitalized in the Pediatric Gastroenterology Ward of Shengjing Hospital affiliated with China Medical University between July 1, 2013, and May 31, 2020, and 20 children who had normal colonoscopy results during the same period (control group) made up the study sample. The disease activity of UC was evaluated based on the pediatric ulcerative colitis activity index, and the ulcerative colitis endoscopic index was evaluated according to the Rachmilewitz score. Immunohistochemical staining was employed to examine the differences in AGR2 expression in the intestinal mucosa between groups. The protective effect of AGR2 in a model of tumor necrosis factor-alpha- (TNF-α-) induced intestinal mucosal barrier injury and the underlying molecular mechanism were explored through in vitro experiments. The results showed that compared with the normal control group, UC patients in the remission or active period had significantly higher expression of AGR2 in the intestine. AGR2 expression was positively correlated with Ki67, an intestinal epithelial cell proliferation marker, but negatively correlated with the degree of endoscopic mucosal injury. In an in vitro model, AGR2 overexpression promoted cell proliferation and migration and inhibited TNF-α-induced intestinal epithelial barrier damage by activating yes-associated protein (YAP). Collectively, our study suggests that AGR2 might serve as a valuable biomarker to help assess the condition and mucosal healing status of UC patients. In vitro, AGR2 promoted the repair of intestinal mucosal barrier injury by activating YAP.
Collapse
|
199
|
Francisco J, Zhang Y, Nakada Y, Jeong JI, Huang CY, Ivessa A, Oka S, Babu GJ, Del Re DP. AAV-mediated YAP expression in cardiac fibroblasts promotes inflammation and increases fibrosis. Sci Rep 2021; 11:10553. [PMID: 34006931 PMCID: PMC8131354 DOI: 10.1038/s41598-021-89989-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
Fibrosis is a hallmark of heart disease independent of etiology and is thought to contribute to impaired cardiac dysfunction and development of heart failure. However, the underlying mechanisms that regulate the differentiation of fibroblasts to myofibroblasts and fibrotic responses remain incompletely defined. As a result, effective treatments to mitigate excessive fibrosis are lacking. We recently demonstrated that the Hippo pathway effector Yes-associated protein (YAP) is an important mediator of myofibroblast differentiation and fibrosis in the infarcted heart. Yet, whether YAP activation in cardiac fibroblasts is sufficient to drive fibrosis, and how fibroblast YAP affects myocardial inflammation, a significant component of adverse cardiac remodeling, are largely unknown. In this study, we leveraged adeno-associated virus (AAV) to target cardiac fibroblasts and demonstrate that chronic YAP expression upregulated indices of fibrosis and inflammation in the absence of additional stress. YAP occupied the Ccl2 gene and promoted Ccl2 expression, which was associated with increased macrophage infiltration, pro-inflammatory cytokine expression, collagen deposition, and cardiac dysfunction in mice with cardiac fibroblast-targeted YAP overexpression. These results are consistent with other recent reports and extend our understanding of YAP function in modulating fibrotic and inflammatory responses in the heart.
Collapse
Affiliation(s)
- Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Yu Zhang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Yasuki Nakada
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Jae Im Jeong
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Andreas Ivessa
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA
| | - Dominic P Del Re
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Avenue, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|
200
|
Wang D, Zhang Y, Xu X, Wu J, Peng Y, Li J, Luo R, Huang L, Liu L, Yu S, Zhang N, Lu B, Zhao K. YAP promotes the activation of NLRP3 inflammasome via blocking K27-linked polyubiquitination of NLRP3. Nat Commun 2021; 12:2674. [PMID: 33976226 PMCID: PMC8113592 DOI: 10.1038/s41467-021-22987-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
The transcription coactivator YAP plays a vital role in Hippo pathway for organ-size control and tissue homeostasis. Recent studies have demonstrated YAP is closely related to immune disorders and inflammatory diseases, but the underlying mechanisms remain less defined. Here, we find that YAP promotes the activation of NLRP3 inflammasome, an intracellular multi-protein complex that orchestrates host immune responses to infections or sterile injuries. YAP deficiency in myeloid cells significantly attenuates LPS-induced systemic inflammation and monosodium urate (MSU) crystals-induced peritonitis. Mechanistically, YAP physically interacts with NLRP3 and maintains the stability of NLRP3 through blocking the association between NLRP3 and the E3 ligase β-TrCP1, the latter increases the proteasomal degradation of NLRP3 via K27-linked ubiquitination at lys380. Together, these findings establish a role of YAP in the activation of NLRP3 inflammasome, and provide potential therapeutic target to treat the NLRP3 inflammasome-related diseases.
Collapse
Affiliation(s)
- Dan Wang
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
- Department of Dermatology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Yening Zhang
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Xueming Xu
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Jianfeng Wu
- State Key Laboratory of Cellular Stress Biology Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Yue Peng
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Jing Li
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Ruiheng Luo
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Lingmin Huang
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Liping Liu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Songlin Yu
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
- Postdoctoral Research Station of Clinical Medicine, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Ningjie Zhang
- Department of Blood Transfusion, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China
| | - Ben Lu
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China.
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan Province, People's Republic of China.
- Key Laboratory of Sepsis and Translational Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan Province, People's Republic of China.
| | - Kai Zhao
- Department of Hematology and Key Laboratory of Non-resolving Inflammation and Cancer of Hunan Province, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, People's Republic of China.
| |
Collapse
|