1
|
Tao X, Wu Y, Guo F, Lv L, Zhai X, Shang D, Yu Z, Xiang H, Dong D. Targeted inhibitors of S100A9 alleviate chronic pancreatitis by inhibiting M2 macrophage polarization via the TAOK3-JNK signaling pathway. Front Immunol 2025; 16:1526813. [PMID: 40207228 PMCID: PMC11979270 DOI: 10.3389/fimmu.2025.1526813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Background Chronic pancreatitis (CP) is a fibro-inflammatory syndrome with unclear pathogenesis and futile therapy. CP's microenvironment disrupts the fine-tuned balance of macrophage polarization toward a predominance of the M2-like phenotype associated with fibrosis. S100A9 is mainly expressed in monocytes as a potent regulator of macrophage phenotype and function. Here, we investigated the S100A9-related mechanisms underlying CP pathology induced by macrophages polarization. Methods S100a9 knockout (S100a9 -/-) mice and an in vitro coculture system of macrophages overexpressing S100a9 and primary PSCs were constructed to investigate the effects and mechanisms of S100A9-mediated macrophage polarization on pancreatic inflammation and fibrosis underpinning CP pathology. Furthermore, a variety of S100A9-targeted small-molecule compounds were screened from U.S. Food and Drug Administration (FDA)-listed drug libraries through molecular docking and virtual screening techniques. Results In CP progression, S100A9 upregulation induces M2 macrophage polarization to accelerate fibrosis via thousand-and-one amino acid kinase 3 (TAOK3)-c-Jun N-terminal kinase (JNK) signaling pathway, and loss of S100A9 reduces CP injury in vitro and in vivo. Coimmunoprecipitation (co-IP) and molecular docking experiments proved that S100A9 may interact directly with TAOK3 through salt bridges and hydrogen bonding interactions of the residues in the S100A9 protein. Furthermore, cobamamide and daptomycin, as inactivators of the S100A9-TAOK3 interaction, can improve CP by inhibiting the polarization of M2 macrophages. Conclusions S100A9 is a significant promoter of M2-like macrophage-induced fibrosis in CP via the TAOK3-JNK signaling pathway. Cobamamide and daptomycin, targeted inhibitors of the S100A9-TAOK3 interaction, may become candidate drugs for CP immunotherapy.
Collapse
Affiliation(s)
- Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Wu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| | - Fangyue Guo
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Linlin Lv
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaohan Zhai
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
- Department of General Surgery, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhan Yu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hong Xiang
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
- Laboratory of Integrative Medicine, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
2
|
Zhang QQ, Ma MM, Chen ZY, Guo YX, Liu K, Xie ML, Wang YL, Li SS, Qian H, Zhang XF, Fu L, Jiang YL. Associations of Serum Legumain with Severity and Prognosis Among Acute Exacerbation of Chronic Obstructive Pulmonary Disease Patients. Int J Chron Obstruct Pulmon Dis 2025; 20:437-447. [PMID: 40027200 PMCID: PMC11871913 DOI: 10.2147/copd.s507018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 02/17/2025] [Indexed: 03/05/2025] Open
Abstract
Background A number of studies have demonstrated that legumain is engaged in the pulmonary diseases. Nevertheless, the role of legumain is indistinct in patients with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). The aim is to identify the correlation of serum legumain with AECOPD patients through a prospective cohort study. Methods All 202 patients with AECOPD were enrolled. Fasting venous blood was collected. Serum legumain was detected by ELISA. Results On admission, serum legumain concentration was gradually elevated in line with AECOPD severity scores. Additionally, serum legumain was closely associated with clinical characteristics. Linear regression analysis confirmed the positive relationships of serum legumain with COPD severity scores. Moreover, the poor prognoses were tracked in patients of AECOPD. Serum higher legumain at admission increased the risks of death and acute exacerbation during hospitalization. Conclusion Serum legumain at admission was positively correlated with the severity and adverse prognosis in AECOPD patients, indicating that legumain plays a vital role in the initiation and development of AECOPD. As a result, serum legumain can become a biomarker in the disease assessment and prognosis prediction for AECOPD.
Collapse
Affiliation(s)
- Qing-Qing Zhang
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Meng-Meng Ma
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Zi-Yong Chen
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Yong-Xia Guo
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Kui Liu
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Mei-Ling Xie
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
- Bengbu Medical University Graduate School, Bengbu, Anhui, 233030, People’s Republic of China
| | - Ying-Li Wang
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
- Bengbu Medical University Graduate School, Bengbu, Anhui, 233030, People’s Republic of China
| | - Shu-Shu Li
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Hui Qian
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Xiao-Fei Zhang
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| | - Lin Fu
- Department of Respiratory and Critical Care Medicine, second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
- Center for Big Data and Population Health of IHM, the second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, People’s Republic of China
| | - Ya-Lin Jiang
- Department of Respiratory and Critical Care Medicine, the Affiliated Bozhou Hospital of Anhui Medical University, Bozhou, Anhui, 236800, People’s Republic of China
| |
Collapse
|
3
|
Ziegler AR, Anderson BM, Latorre R, McQuade RM, Dufour A, Schmidt BL, Bunnett NW, Scott NE, Edgington‐Mitchell LE. N-terminomics profiling of naïve and inflamed murine colon reveals proteolytic signatures of legumain. J Cell Physiol 2025; 240:e31466. [PMID: 39392222 PMCID: PMC11735880 DOI: 10.1002/jcp.31466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/27/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024]
Abstract
Legumain is a cysteine protease broadly associated with inflammation. It has been reported to cleave and activate protease-activated receptor 2 to provoke pain associated with oral cancer. Outside of gastric and colon cancer, little has been reported on the roles of legumain within the gastrointestinal tract. Using a legumain-selective activity-based probe, LE28, we report that legumain is activated within colonocytes and macrophages of the murine colon, and that it is upregulated in models of acute experimental colitis. We demonstrated that loss of legumain activity in colonocytes, either through pharmacological inhibition or gene deletion, had no impact on epithelial permeability in vitro. Moreover, legumain inhibition or deletion had no obvious impacts on symptoms or histological features associated with dextran sulfate sodium-induced colitis, suggesting its proteolytic activity is dispensable for colitis initiation. To gain insight into potential functions of legumain within the colon, we performed field asymmetric waveform ion mobility spectrometry-facilitated quantitative proteomics and N-terminomics analyses on naïve and inflamed colon tissue from wild-type and legumain-deficient mice. We identified 16 altered cleavage sites with an asparaginyl endopeptidase signature that may be direct substrates of legumain and a further 16 cleavage sites that may be indirectly mediated by legumain. We also analyzed changes in protein abundance and proteolytic events broadly associated with colitis in the gut, which permitted comparison to recent analyses on mucosal biopsies from patients with inflammatory bowel disease. Collectively, these results shed light on potential functions of legumain and highlight its potential roles in the transition from inflammation to colorectal cancer.
Collapse
Affiliation(s)
- Alexander R. Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Bethany M. Anderson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Rocco Latorre
- Department of Molecular PathobiologyNew York University College of DentistryNew YorkNew YorkUSA
| | - Rachel M. McQuade
- Department of Anatomy and PhysiologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Antoine Dufour
- Department of Physiology and PharmacologyUniversity of CalgaryCalgaryAlbertaCanada
| | - Brian L. Schmidt
- Department of Oral and Maxillofacial SurgeryNew York University College of Dentistry, Bluestone Center for Clinical ResearchNew YorkNew YorkUSA
| | - Nigel W. Bunnett
- Department of Molecular PathobiologyNew York University College of DentistryNew YorkNew YorkUSA
| | - Nichollas E. Scott
- Department of Microbiology and Immunology, Peter Doherty InstituteThe University of MelbourneParkvilleVictoriaAustralia
| | - Laura E. Edgington‐Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology InstituteThe University of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
4
|
Zhang W, Wang S, Liu Z, Qian P, Li Y, Wu J. Legumain-deficient macrophages regulate inflammation and lipid metabolism in adipose tissues to protect against diet-induced obesity. Mol Cell Endocrinol 2024; 592:112283. [PMID: 38815795 DOI: 10.1016/j.mce.2024.112283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/01/2024]
Abstract
Adipose tissue macrophages (ATMs) are key players in the development of obesity and associated metabolic inflammation, which contributes to systemic metabolic dysfunction, and understanding the interaction between macrophages and adipocytes is crucial for developing novel macrophage-based strategies against obesity. Here, we found that Legumain (Lgmn), a well-known lysosomal cysteine protease, is expressed mainly in the ATMs of obese mice. To further define the potential role of Lgmn-expressing macrophages in the generation of an aberrant metabolic state, LgmnF/F; LysMCre mice, which do not express Lgmn in macrophages, were maintained on a high-fat diet (HFD), and metabolic parameters were assessed. Macrophage-specific Lgmn deficiency protects mice against HFD-induced obesity, diminishes the quantity of proinflammatory macrophages in obese adipose tissues, and alleviates hepatic steatosis and insulin resistance. By analysing the transcriptome and proteome of murine visceral white adipose tissue (vWAT) after HFD feeding, we determined that macrophage Lgmn deficiency causes changes in lipid metabolism and the inflammatory response. Furthermore, the reciprocity of macrophage-derived Lgmn with integrin α5β1 in adipocytes was tested via colocalization analyses. It is further demonstrated in macrophage and adipocyte coculture system that macrophage derived Lgmn bound to integrin α5β1 in adipocytes, therefore attenuating PKA activation, downregulating lipolysis-related proteins and eventually exacerbating obesity development. Overall, our study identified Lgmn as a previously unrecognized regulator involved in the interaction between ATMs and adipocytes contributing to diet-induced obesity and suggested that Lgmn is a potential target for treating metabolic disorders.
Collapse
Affiliation(s)
- Wanyu Zhang
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China
| | - Shuowen Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zhuo Liu
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Ping Qian
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Yuanyuan Li
- Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China
| | - Jianxin Wu
- Children's Hospital Capital Institute of Pediatrics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Department of Biochemistry and Immunology, Capital Institute of Pediatrics, Beijing, China; Graduate School of Peking Union Medical College, Beijing, China; Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
5
|
Ziegler AR, Dufour A, Scott NE, Edgington-Mitchell LE. Ion Mobility-Based Enrichment-Free N-Terminomics Analysis Reveals Novel Legumain Substrates in Murine Spleen. Mol Cell Proteomics 2024; 23:100714. [PMID: 38199506 PMCID: PMC10862022 DOI: 10.1016/j.mcpro.2024.100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/12/2024] Open
Abstract
Aberrant levels of the asparaginyl endopeptidase legumain have been linked to inflammation, neurodegeneration, and cancer, yet our understanding of this protease is incomplete. Systematic attempts to identify legumain substrates have been previously confined to in vitro studies, which fail to mirror physiological conditions and obscure biologically relevant cleavage events. Using high-field asymmetric waveform ion mobility spectrometry (FAIMS), we developed a streamlined approach for proteome and N-terminome analyses without the need for N-termini enrichment. Compared to unfractionated proteomic analysis, we demonstrate FAIMS fractionation improves N-termini identification by >2.5 fold, resulting in the identification of >2882 unique N-termini from limited sample amounts. In murine spleens, this approach identifies 6366 proteins and 2528 unique N-termini, with 235 cleavage events enriched in WT compared to legumain-deficient spleens. Among these, 119 neo-N-termini arose from asparaginyl endopeptidase activities, representing novel putative physiological legumain substrates. The direct cleavage of selected substrates by legumain was confirmed using in vitro assays, providing support for the existence of physiologically relevant extra-lysosomal legumain activity. Combined, these data shed critical light on the functions of legumain and demonstrate the utility of FAIMS as an accessible method to improve depth and quality of N-terminomics studies.
Collapse
Affiliation(s)
- Alexander R Ziegler
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Dufour
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Nichollas E Scott
- Department of Microbiology and Immunology, Peter Doherty Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Pang L, Guo S, Khan F, Dunterman M, Ali H, Liu Y, Huang Y, Chen P. Hypoxia-driven protease legumain promotes immunosuppression in glioblastoma. Cell Rep Med 2023; 4:101238. [PMID: 37858339 PMCID: PMC10694605 DOI: 10.1016/j.xcrm.2023.101238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/27/2023] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
Glioblastoma (GBM) is a hypoxic and "immune-cold" tumor containing rich stromal signaling molecules and cell populations, such as proteases and immunosuppressive tumor-associated macrophages (TAMs). Here, we seek to profile and characterize the potential proteases that may contribute to GBM immunosuppression. Legumain (LGMN) emerges as the key protease that is highly enriched in TAMs and transcriptionally upregulated by hypoxia-inducible factor 1-alpha (HIF1α). Functionally, the increased LGMN promotes TAM immunosuppressive polarization via activating the GSK-3β-STAT3 signaling pathway. Inhibition of macrophage HIF1α and LGMN reduces TAM immunosuppressive polarization, impairs tumor progression, enhances CD8+ T cell-mediated anti-tumor immunity, and synergizes with anti-PD1 therapy in GBM mouse models. Thus, LGMN is a key molecular switch connecting two GBM hallmarks of hypoxia and immunosuppression, providing an actionable therapeutic intervention for this deadly disease.
Collapse
Affiliation(s)
- Lizhi Pang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Songlin Guo
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Fatima Khan
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Madeline Dunterman
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Heba Ali
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yang Liu
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yuyun Huang
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Peiwen Chen
- Department of Neurological Surgery, Lou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Ni YH, Wang R, Wang W, Li DZ, Liu G, Jiang CS, Wang Y, Lin X, Zeng XP. Tcf21 Alleviates Pancreatic Fibrosis by Regulating the Epithelial-Mesenchymal Transformation of Pancreatic Stellate Cells. Dig Dis Sci 2023:10.1007/s10620-023-07849-w. [PMID: 36943591 DOI: 10.1007/s10620-023-07849-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 01/25/2023] [Indexed: 03/23/2023]
Abstract
BACKGROUND AND AIMS The activation of pancreatic stellate cells (PSCs) plays a key role in the occurrence and development of chronic pancreatitis (CP) and pancreatic fibrosis, which is related to the process of epithelial-mesenchymal transition (EMT). This study was designed to investigate the effect and mechanism of Tcf21 (one of tumor suppressor genes) on pancreatic inflammation and fibrosis in vivo and in vitro. METHODS C57BL/6 male mice were intraperitoneally injected with caerulein for 6 weeks to establish CP animal model. Fixed pancreatic tissue paraffin-embedded sections were used for immunohistochemistry staining of Tcf21, fibrosis-related markers (α-SMA), interstitial markers (Vimentin) and epithelial markers (E-cadherin). Western blotting and qRT-PCR assay were performed to analyze the change of expression of the above markers after stimulation of TGF-β1 or overexpressed Tcf21 lentivirus transfection in human pancreatic stellate cells (HPSCs). RESULTS The pancreatic expression of α-SMA and Vimentin of CP mice significantly increased, while the expression of Tcf21 and E-cadherin significantly decreased. TGF-β1 could promote activation and EMT process of HPSCs, and inhibited the expression of Tcf21. Overexpression of Tcf21 could significantly down-regulate the expression of α-SMA, Fibronectin and Vimentin, and up-regulated the expression of ZO-1 of HPSCs. Cell Counting Kit-8 assay and scratch wound-healing assay results showed that overexpression of Tcf21 could significantly inhibit the cell migration and proliferation of HPSCs. CONCLUSIONS Overexpression of Tcf21 could significantly alleviate the activation, proliferation, migration of PSCs by regulating the EMT process. Tcf21 had a potential prospect of a new target for CP therapy.
Collapse
Affiliation(s)
- Yan-Hong Ni
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Rong Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Wen Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Da-Zhou Li
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Gang Liu
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Chuan-Shen Jiang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China
| | - Yi Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xia Lin
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China
| | - Xiang-Peng Zeng
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China.
- Department of Digestive Diseases, Fuzong Clinical Medical College, Fujian Medical University, 156 North Road of West No.2 Ring, Fuzhou, 350025, China.
- Department of Digestive Diseases, Dongfang Hospital, Xiamen University, Fuzhou, China.
| |
Collapse
|
8
|
Solberg R, Lunde NN, Forbord KM, Okla M, Kassem M, Jafari A. The Mammalian Cysteine Protease Legumain in Health and Disease. Int J Mol Sci 2022; 23:ijms232415983. [PMID: 36555634 PMCID: PMC9788469 DOI: 10.3390/ijms232415983] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/23/2022] Open
Abstract
The cysteine protease legumain (also known as asparaginyl endopeptidase or δ-secretase) is the only known mammalian asparaginyl endopeptidase and is primarily localized to the endolysosomal system, although it is also found extracellularly as a secreted protein. Legumain is involved in the regulation of diverse biological processes and tissue homeostasis, and in the pathogenesis of various malignant and nonmalignant diseases. In addition to its proteolytic activity that leads to the degradation or activation of different substrates, legumain has also been shown to have a nonproteolytic ligase function. This review summarizes the current knowledge about legumain functions in health and disease, including kidney homeostasis, hematopoietic homeostasis, bone remodeling, cardiovascular and cerebrovascular diseases, fibrosis, aging and senescence, neurodegenerative diseases and cancer. In addition, this review addresses the effects of some marketed drugs on legumain. Expanding our knowledge on legumain will delineate the importance of this enzyme in regulating physiological processes and disease conditions.
Collapse
Affiliation(s)
- Rigmor Solberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| | - Ngoc Nguyen Lunde
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
| | - Karl Martin Forbord
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, N-0316 Oslo, Norway
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
| | - Meshail Okla
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Abbas Jafari
- Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, DK-5000 Odense, Denmark
- Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Correspondence: (R.S.); (A.J.); Tel.: +47-22-857-514 (R.S.); +45-35-337-423 (A.J.)
| |
Collapse
|
9
|
Wendisch D, Dietrich O, Mari T, von Stillfried S, Ibarra IL, Mittermaier M, Mache C, Chua RL, Knoll R, Timm S, Brumhard S, Krammer T, Zauber H, Hiller AL, Pascual-Reguant A, Mothes R, Bülow RD, Schulze J, Leipold AM, Djudjaj S, Erhard F, Geffers R, Pott F, Kazmierski J, Radke J, Pergantis P, Baßler K, Conrad C, Aschenbrenner AC, Sawitzki B, Landthaler M, Wyler E, Horst D, Hippenstiel S, Hocke A, Heppner FL, Uhrig A, Garcia C, Machleidt F, Herold S, Elezkurtaj S, Thibeault C, Witzenrath M, Cochain C, Suttorp N, Drosten C, Goffinet C, Kurth F, Schultze JL, Radbruch H, Ochs M, Eils R, Müller-Redetzky H, Hauser AE, Luecken MD, Theis FJ, Conrad C, Wolff T, Boor P, Selbach M, Saliba AE, Sander LE. SARS-CoV-2 infection triggers profibrotic macrophage responses and lung fibrosis. Cell 2021; 184:6243-6261.e27. [PMID: 34914922 PMCID: PMC8626230 DOI: 10.1016/j.cell.2021.11.033] [Citation(s) in RCA: 337] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 07/28/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
COVID-19-induced "acute respiratory distress syndrome" (ARDS) is associated with prolonged respiratory failure and high mortality, but the mechanistic basis of lung injury remains incompletely understood. Here, we analyze pulmonary immune responses and lung pathology in two cohorts of patients with COVID-19 ARDS using functional single-cell genomics, immunohistology, and electron microscopy. We describe an accumulation of CD163-expressing monocyte-derived macrophages that acquired a profibrotic transcriptional phenotype during COVID-19 ARDS. Gene set enrichment and computational data integration revealed a significant similarity between COVID-19-associated macrophages and profibrotic macrophage populations identified in idiopathic pulmonary fibrosis. COVID-19 ARDS was associated with clinical, radiographic, histopathological, and ultrastructural hallmarks of pulmonary fibrosis. Exposure of human monocytes to SARS-CoV-2, but not influenza A virus or viral RNA analogs, was sufficient to induce a similar profibrotic phenotype in vitro. In conclusion, we demonstrate that SARS-CoV-2 triggers profibrotic macrophage responses and pronounced fibroproliferative ARDS.
Collapse
Affiliation(s)
- Daniel Wendisch
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Oliver Dietrich
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Tommaso Mari
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Proteome Dynamics, Berlin, Germany
| | | | - Ignacio L Ibarra
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Mirja Mittermaier
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Christin Mache
- Unit 17 Influenza and other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Robert Lorenz Chua
- Center for Digital Health, Berlin Institute of Health (BIH) and Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rainer Knoll
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany; Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Sara Timm
- Core Facility Electron Microscopy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Sophia Brumhard
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Krammer
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Henrik Zauber
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Proteome Dynamics, Berlin, Germany
| | - Anna Luisa Hiller
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Pascual-Reguant
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charité; Deutsches Rheumaforschungszentrum, Immunodynamics, a Leibniz Institute, Berlin, Germany
| | - Ronja Mothes
- Deutsches Rheumaforschungszentrum, Immunodynamics, a Leibniz Institute, Berlin, Germany; Charité - Universitätsmedizin Berlin, Department of Neuropathology, Berlin, Germany
| | - Roman David Bülow
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Jessica Schulze
- Unit 17 Influenza and other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Alexander M Leipold
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Sonja Djudjaj
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Florian Erhard
- Institute for Virology and Immunobiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Robert Geffers
- Genome Analytics, Helmholtz-Center for Infection Research (HZI), Braunschweig, Germany
| | - Fabian Pott
- Berlin Institute of Health (BIH), Berlin, Germany; Charité - Universitätsmedizin Berlin, Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Kazmierski
- Berlin Institute of Health (BIH), Berlin, Germany; Charité - Universitätsmedizin Berlin, Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Josefine Radke
- Berlin Institute of Health (BIH), Berlin, Germany; Charité - Universitätsmedizin Berlin, Department of Neuropathology, Berlin, Germany
| | - Panagiotis Pergantis
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Kevin Baßler
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany; Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Claudia Conrad
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Anna C Aschenbrenner
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany; Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), and University of Bonn, Bonn, Germany; Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Birgit Sawitzki
- Charité, Universitätsmedizin Berlin, Institute of Medical Immunology, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Landthaler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Emanuel Wyler
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - David Horst
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Stefan Hippenstiel
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany
| | - Andreas Hocke
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany
| | - Frank L Heppner
- Charité - Universitätsmedizin Berlin, Department of Neuropathology, Berlin, Germany; Cluster of Excellence, NeuroCure, Berlin, Germany; German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
| | - Alexander Uhrig
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Garcia
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Machleidt
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Herold
- German Center for Lung Research (DZL), Germany; Division of Infectious Diseases, Pulmonary and Critical Care Medicine, Department of Internal Medicine II, Universities of Giessen and Marburg Lung Center, Giessen, Germany; Institute of Lung Health (ILH), Giessen, Germany
| | - Sefer Elezkurtaj
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Charlotte Thibeault
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany
| | - Clément Cochain
- Comprehensive Heart Failure Center Wuerzburg, University Hospital Würzburg, Germany; Institute of Experimental Biomedicine, University Hospital Würzburg, Würzburg, Germany
| | - Norbert Suttorp
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany
| | - Christian Drosten
- Charité - Universitätsmedizin Berlin, Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany; German Center for Infection Research (DZIF), Braunschweig, Germany
| | - Christine Goffinet
- Berlin Institute of Health (BIH), Berlin, Germany; Charité - Universitätsmedizin Berlin, Institute of Virology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Kurth
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany; I. Department of Medicine, University Medical Center, Hamburg-Eppendorf, Hamburg, Germany
| | - Joachim L Schultze
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany; Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), and University of Bonn, Bonn, Germany
| | - Helena Radbruch
- Charité - Universitätsmedizin Berlin, Department of Neuropathology, Berlin, Germany
| | - Matthias Ochs
- Core Facility Electron Microscopy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany; Institute of Functional Anatomy, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health (BIH) and Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Holger Müller-Redetzky
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany
| | - Anja E Hauser
- Charité - Universitätsmedizin Berlin, Department of Rheumatology and Clinical Immunology, Charité; Deutsches Rheumaforschungszentrum, Immunodynamics, a Leibniz Institute, Berlin, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany; Department of Mathematics, Technische Universität München, Garching bei München, Germany
| | - Christian Conrad
- Center for Digital Health, Berlin Institute of Health (BIH) and Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thorsten Wolff
- Unit 17 Influenza and other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Matthias Selbach
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Proteome Dynamics, Berlin, Germany; Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany.
| | - Leif Erik Sander
- Charité - Universitätsmedizin Berlin, Department of Infectious Diseases and Respiratory Medicine, Charité, Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Germany.
| |
Collapse
|
10
|
Wu D, Guo J, Qi B, Xiao H. TGF-β1 induced proliferation, migration, and ECM accumulation through the SNHG11/miR-34b/LIF pathway in human pancreatic stellate cells. Endocr J 2021; 68:1347-1357. [PMID: 34261825 DOI: 10.1507/endocrj.ej21-0176] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chronic pancreatitis (CP) is a chronic inflammatory and fibrotic disease of the pancreas, and activated pancreatic stellate cells (PSCs) play a vital role in the progression of pancreatic fibrosis in CP. It has been reported that long non-coding RNA small nucleolar RNA host gene 11 (SNHG11) is highly expressed in chronic pancreatitis (CP) patients. However, the role of SNHG11 in CP progression is unclear. The purport of the study was to survey the role of SNHG11 in CP. We employed transforming growth factor (TGF)-beta1 (TGF-β1) to activate human pancreatic stellate cells (PSCs). Expression of SNHG11 was assessed with qRT-PCR. Loss-of-function experiments were executed to evaluate the effects of SNHG11 on the proliferation and migration of TGF-β1-treated PSCs. Some protein levels were detected by western blotting. The regulatory mechanism of SNHG11 was verified by the dual-luciferase reporter and RIP assays. As a result, SNHG11 was upregulated in plasma of CP patients and TGF-β1-treated PSCs. Also, SNHG11 inhibition reduced TGF-β1-induced proliferation, migration, and ECM accumulation in PSCs. Mechanistically, SNHG11 regulated leukemia inhibitory factor (LIF) expression by sponging miR-34b. Furthermore, miR-34b inhibitor abolished SNHG11 silencing-mediated effects on TGF-β1-treated PSC proliferation, migration, and ECM accumulation. LIF overexpression counteracted the repressive influence of miR-34b mimic on proliferation, migration, and ECM accumulation of TGF-β1-treated PSCs. In conclusion, SNHG11 knockdown reduced TGF-β1-induced PSC proliferation, migration, and ECM accumulation by the miR-34b/LIF axis.
Collapse
Affiliation(s)
- Desheng Wu
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Jin Guo
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Benquan Qi
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| | - Heng Xiao
- Department of Emergency Medicine, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233000, China
| |
Collapse
|
11
|
Sofias AM, De Lorenzi F, Peña Q, Azadkhah Shalmani A, Vucur M, Wang JW, Kiessling F, Shi Y, Consolino L, Storm G, Lammers T. Therapeutic and diagnostic targeting of fibrosis in metabolic, proliferative and viral disorders. Adv Drug Deliv Rev 2021; 175:113831. [PMID: 34139255 PMCID: PMC7611899 DOI: 10.1016/j.addr.2021.113831] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023]
Abstract
Fibrosis is a common denominator in many pathologies and crucially affects disease progression, drug delivery efficiency and therapy outcome. We here summarize therapeutic and diagnostic strategies for fibrosis targeting in atherosclerosis and cardiac disease, cancer, diabetes, liver diseases and viral infections. We address various anti-fibrotic targets, ranging from cells and genes to metabolites and proteins, primarily focusing on fibrosis-promoting features that are conserved among the different diseases. We discuss how anti-fibrotic therapies have progressed over the years, and how nanomedicine formulations can potentiate anti-fibrotic treatment efficacy. From a diagnostic point of view, we discuss how medical imaging can be employed to facilitate the diagnosis, staging and treatment monitoring of fibrotic disorders. Altogether, this comprehensive overview serves as a basis for developing individualized and improved treatment strategies for patients suffering from fibrosis-associated pathologies.
Collapse
Affiliation(s)
- Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO(ABCD)), University Hospital Aachen, Aachen, Germany; Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Quim Peña
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Armin Azadkhah Shalmani
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Duesseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Fabian Kiessling
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Yang Shi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Lorena Consolino
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, the Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Aachen, Germany; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands; Department of Targeted Therapeutics, University of Twente, Enschede, the Netherlands.
| |
Collapse
|
12
|
Zeng XP, Zeng JH, Lin X, Ni YH, Jiang CS, Li DZ, He XJ, Wang R, Wang W. Puerarin Ameliorates Caerulein-Induced Chronic Pancreatitis via Inhibition of MAPK Signaling Pathway. Front Pharmacol 2021; 12:686992. [PMID: 34149430 PMCID: PMC8207514 DOI: 10.3389/fphar.2021.686992] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/18/2021] [Indexed: 01/14/2023] Open
Abstract
Pancreatic fibrosis is one of the most important pathological features of chronic pancreatitis (CP), and pancreatic stellate cells (PSCs) are considered to be the key cells. Puerarin is the most important flavonoid active component in Chinese herb Radix Puerariae, and it exhibited anti-fibrotic effect in various fibrous diseases recently. However, the impact and molecular mechanism of puerarin on CP and pancreatic fibrosis remain unknown. This study systematically investigated the effect of puerarin on CP and pancreatic fibrosis in vivo and in vitro. H&E staining, Sirius Red staining, qRT-PCR and Western blotting analysis of fibrosis and inflammation related genes of pancreatic tissues showed that puerarin notably ameliorated pancreatic atrophy, inflammation and fibrosis in a model of caerulein-induced murine CP. Western blotting analysis of pancreatic tissues showed the phosphorylation level of MAPK family proteins (JNK1/2, ERK1/2 and p38 MAPK) significantly increased after modeling of cerulein, while puerarin could inhibit their phosphorylation levels to a certain extent. We found that puerarin exerted a marked inhibition on the proliferation, migration and activation of PSCs, determined by CCK-8 assay, transwell migration assay, scratch wound-healing assay and expression levels of α-SMA, Fibronectin, Col1α1 and GFAP. Western blotting result demonstrated that puerarin markedly inhibited the phosphorylation of MAPK family proteins (JNK1/2, ERK1/2 and p38 MAPK) of PSCs in a dose-dependent manner whether or not stimulated by platelet-activating factor. In conclusion, the present study showed that puerarin could be a potential therapeutic candidate in the treatment of CP, and the MAPK pathway might be its important target.
Collapse
Affiliation(s)
- Xiang-Peng Zeng
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Jing-Hui Zeng
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Xia Lin
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Yan-Hong Ni
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Chuan-Shen Jiang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Da-Zhou Li
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Xiao-Jian He
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Rong Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Wen Wang
- Department of Digestive Diseases, 900TH Hospital of Joint Logistics Support Force, Fuzhou General Clinical Medical College of Fujian Medical University, Oriental Hospital Affiliated to Xiamen University, Fuzhou, China
| |
Collapse
|