1
|
Ban J, Qian J, Zhang C, Li J. Recent advances in TAM mechanisms in lung diseases. J Transl Med 2025; 23:479. [PMID: 40287707 PMCID: PMC12032715 DOI: 10.1186/s12967-025-06398-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/18/2025] [Indexed: 04/29/2025] Open
Abstract
TYRO3, MERTK, and AXL receptor tyrosine kinases, collectively known as TAM receptors, play a vital role in maintaining lung tissue homeostasis by regulating integrity and self-renewal. These receptors activate signalling pathways that inhibit apoptosis, promote cell proliferation and differentiation, mediate cell adhesion and migration, and perform other essential biological functions. Additionally, TAM receptors are implicated in mechanisms that suppress anti-tumor immunity and confer resistance to immune checkpoint inhibitors. Disruption of the homeostatic balances can lead to pathological conditions such as lung inflammation, fibrosis, or tumors. Recent studies highlight their significant role in COVID-19-induced lung injury. However, the exact mechanisms by which TAM receptors contribute to lung diseases remain unclear. This article reviews the potential mechanisms of TAM receptor involvement in disease progression, focusing on lung inflammation, fibrosis, cancer, and COVID-19-induced lung injury. It also explores future research aspects and the therapeutic potentials of targeting TAM receptors, providing a theoretical foundation for understanding lung disease mechanisms and identifying treatment targets.
Collapse
Affiliation(s)
- Jiaqi Ban
- School of Public Health, The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, China
| | - Jiayi Qian
- School of Public Health, The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, China
| | - Chi Zhang
- School of Clinical Medicine, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, 561113, People's Republic of China
| | - Jun Li
- School of Public Health, The key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guiyang, 561113, Guizhou, China.
| |
Collapse
|
2
|
Li J, Ren X, Cao L, Min K, Wang M, Lin S, Lei L, Tan Z, Cheng X, Li X, Guan F. Bisecting GlcNAc modification of vesicular GAS6 regulates CAFs activation and breast cancer metastasis. Cell Commun Signal 2025; 23:39. [PMID: 39844194 PMCID: PMC11755815 DOI: 10.1186/s12964-025-02052-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) are a pivotal component of the tumor microenvironment (TME), playing key roles in tumor initiation, metastasis, and chemoresistance. While glycosylation is known to regulate various cellular processes, its impact on CAFs activation remains insufficiently explored. METHODS We assessed the correlation between bisecting GlcNAc levels and CAFs markers (α-SMA, PDGFRA, PDGFRB) in breast cancer tissues. The effects of small extracellular vesicles (sEVs) derived from MDA-MB-231/OEvec and MDA-MB-231/OEMGAT3 cells on CAFs activation were examined using western blotting, transwell, and collagen contraction assays. Proteomic analysis was performed to identify dysregulated proteins in sEVs from different cell lines. The role of GAS6 in CAFs activation was validated through in vitro and in vivo experiments. The impact of bisecting GlcNAc modification on GAS6 expression and function was analyzed through protein degradation and N-glycosylation site mutation. The effect of activated CAFs on breast cancer metastasis was evaluated using western blotting and transwell assays. RESULTS We found that low bisecting GlcNAc levels were associated with CAFs activation within the TME of breast cancer. Breast cancer-derived sEVs stimulated the conversion of normal fibroblasts to CAFs, with GAS6 in sEVs playing a key role by interacting with AXL receptors on fibroblasts. Introducing GAS6 into normal fibroblasts induced their conversion into CAFs, which enhanced breast cancer cell metastasis. Notably, GAS6 was decorated with bisecting GlcNAc, which promoted its degradation in donor cells, leading to reduced GAS6 levels in sEVs and attenuating GAS6-mediated CAFs activation. CONCLUSION Taken together, our findings provide new insights into the functional role of bisecting GlcNAc on GAS6-mediated CAFs activation in breast cancer.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China
- Xi'an No. 1 Hospital, First Affiliated Hospital of Northwest University, Xi'an, 710002, P. R. China
| | - Xueting Ren
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Lin Cao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China
| | - Kaixiang Min
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China
| | - Meng Wang
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Shuai Lin
- The Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, P. R. China
| | - Lei Lei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China
| | - Zengqi Tan
- Institute of Hematology, School of Medicine, Northwest University, Xi'an, 710069, P. R. China
| | - Xiaoliang Cheng
- Northwest University Qlife Precision Medicine Center, Xi'an, 710069, P. R. China
| | - Xiang Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, 229 Taibai North Road, Xi'an, Shaanxi, 710069, P. R. China.
| |
Collapse
|
3
|
Xu H, Xue S, Sun Y, Ma J, Li S, Wang Y, Mao T, Ge W, Yue M, Shentu D, Lu W, Wang Y, Hu J, Cui J, Zhang X, Cai L, Wang Y, Wang L. CREB3L1 facilitates pancreatic tumor progression and reprograms intratumoral tumor-associated macrophages to shape an immunotherapy-resistance microenvironment. J Immunother Cancer 2025; 13:e010029. [PMID: 39762079 PMCID: PMC11749327 DOI: 10.1136/jitc-2024-010029] [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: 07/09/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND To date, a growing body of evidence suggests that unfolded protein response (UPR) sensors play a vital role in carcinogenesis. However, it remains unclear whether they are involved in pancreatic ductal adenocarcinoma (PDAC) and how they relate to clinical outcomes. This study aims to investigate the biological function and mechanism of how a novel UPR sensor, CREB3L1 works in PDAC and further evaluate its clinical application prospect. METHODS We tested UPR signaling including CREB3L1 in Thapsigargin-treated PDAC cells. Subsequently, we defined CREB3L1 expression and further analyzed its expression with clinical characteristics in PDAC. Then, we established gene-modified cells to determine whether CREB3L1 functions in cell proliferation and migration capacity. Besides, we constructed subcutaneously and orthotopically transplanted mice models to verify their progrowing function and pulmonary metastasis models to prove their proinvasion role. What's more, RNAseq, qPCR, Western blotting, immunohistochemistry and multicolor flow cytometry experiments were used to explore the mechanism of how CREB3L1 worked in PDAC. Lastly, CREB3L1 expression correlation with PDAC immunotherapy outcome and immune cell signatures were explored in the patients with advanced PDAC who received PD-1 antibody therapy. RESULTS We first confirmed CREB3L1 could be induced by endoplasmic reticulum stressor and found its aberrant activation was associated with poorer overall survival in PDAC patients indicating the protumor function of the new UPR sensor. Functionally, we confirmed CREB3L1 contributing to PDAC malignant progression including growth and metastasis by multiple in in vitro and in vivo models. Mechanistically, CREB3L1 upregulated COL3A1 and promoted dense stroma formation for facilitating PDAC and knocking down COL3A1 disrupted CREB3L1 protumor function. Furthermore, CREB3L1-induced TAM polarization toward an M2 phenotype and reduced the infiltration of CD8+ T cells. Clinically, CREB3L1 correlated with immune cell signatures as well as immune checkpoint blockade (ICB) treatment response and outcome that CREB3L1aberrant activation indicated poorer efficacy and worse prognosis than the low in PDAC which might empower clinical decision. CONCLUSIONS Collectively, this study revealed CREB3L1 facilitated PDAC progression, shaped an immune exclude tumor microenvironment and distinguished therapy response and outcome of ICB therapy indicating CREB3L1 could be a promising novel molecular target and biomarker for PDAC treatment.
Collapse
Affiliation(s)
- Haiyan Xu
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shengbai Xue
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Sun
- Institute of Molecular Medicine (IMM), State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyu Ma
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shumin Li
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanling Wang
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tiebo Mao
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weiyu Ge
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Yue
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Daiyuan Shentu
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Lu
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongchao Wang
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiong Hu
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jiujie Cui
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaofei Zhang
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Cai
- Department of Infectious Diseases, Shanghai Sixth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yu Wang
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Liwei Wang
- State Key Laboratory of Systems Medicine for Cancer of Oncology Department and Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Zhao X, Li Y, Yang S, Chen Y, Wu K, Geng J, Liu P, Wang Z, Dai H, Wang C. Orderly Regulation of Macrophages and Fibroblasts by Axl in Bleomycin-Induced Pulmonary Fibrosis in Mice. J Cell Mol Med 2025; 29:e70321. [PMID: 39779468 PMCID: PMC11710931 DOI: 10.1111/jcmm.70321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 11/26/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Pulmonary fibrosis is a pathological manifestation that occurs upon lung injury and subsequence aberrant repair with poor prognosis. However, current treatment is limited and does not distinguish different disease stages. Here, we aimed to study the differential functions of Axl, a receptor tyrosine kinase expressing on both macrophages and fibroblasts, in the whole course of pulmonary fibrosis. We used mice with Axl total knockout, conditionally knockout in macrophages or fibroblasts, or treating with Axl inhibitors in inflammation or fibrosis stages to examine the effect of temporary dysfunction of Axl on bleomycin (BLM)-induced pulmonary fibrosis. Primary bone marrow-derived monocytes and primary fibroblasts from mice were used for cell-type-specific studies. Lung tissue and plasma samples were collected from idiopathic pulmonary fibrosis (IPF) patients and healthy controls to assess the Axl levels. We found that Axl inhibited the M1 polarisation of macrophages; inhibition of Axl during acute phase exacerbated inflammatory response and subsequent pulmonary fibrosis. On the other hand, Axl promoted the proliferation and invasion of the fibroblasts, partially by accelerating the focal adhesion turnover; inhibiting Axl during the fibrotic phase significantly alleviated pulmonary fibrosis. Consistently, phosphorylated Axl levels increased in fibrotic foci in the lung sample of IPF patients. In contrast, the soluble Axl (sAxl) level decreased in their plasma as compared to healthy controls. These results indicate that Axl may sequentially and differentially regulate macrophages and fibroblasts in acute and fibrosis phases, implying the necessity of a stage-specific treatment for pulmonary fibrosis. In addition, the activated Axl on fibroblasts may be reflected by the lowered plasma sAxl level, which may act as a biomarker for IPF. Trial Registration: ClinicalTrials.gov identifier: NCT03730337.
Collapse
Affiliation(s)
- Xinyu Zhao
- The Second Affiliated Hospital of Harbin Medical UniversityHeilongjiangChina
| | - Yupeng Li
- The Second Affiliated Hospital of Harbin Medical UniversityHeilongjiangChina
| | - Shengnan Yang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesChina‐Japan Friendship HospitalBeijingChina
- National Center for Respiratory Medicine, Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
- Department of Respiratory and Critical Care MedicineTianjin Chest HospitalChina
| | | | - Kaiwei Wu
- Peking Union Medical CollegeBeijingChina
| | - Jing Geng
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesChina‐Japan Friendship HospitalBeijingChina
- National Center for Respiratory Medicine, Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
| | - Peipei Liu
- Department of Medicine and Women's Guild Lung InstituteCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Zai Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesChina‐Japan Friendship HospitalBeijingChina
- National Center for Respiratory Medicine, Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
- Institute of Clinical Medical SciencesChina‐Japan Friendship HospitalBeijingChina
| | - Huaping Dai
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesChina‐Japan Friendship HospitalBeijingChina
- National Center for Respiratory Medicine, Institute of Respiratory MedicineChinese Academy of Medical SciencesBeijingChina
| | - Chen Wang
- The Second Affiliated Hospital of Harbin Medical UniversityHeilongjiangChina
| |
Collapse
|
5
|
de Aguiar ACF, Ferreira NCFDL, Borba MACSM, Filho DDLF, Leitão GM, Mattos LA, Filho JLDL, Martins DBG. TYRO3 and EPHA2 Expression Are Dysregulated in Breast Cancer. Cell Biochem Funct 2024; 42:e4128. [PMID: 39327735 DOI: 10.1002/cbf.4128] [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: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024]
Abstract
Receptor tyrosine kinases (RTKs) are involved in cell growth, motility, and differentiation. Deregulation of RTKs signaling is associated with tumor development and therapy resistance. Potential RTKs like TAM (TYRO3, AXL, MERTK), RON, EPH, and MET have been evaluated in many cancers like lung, prostate, and colorectal, but little is known in breast tumors. In this study, 51 luminal breast cancer tissue and 8 triple negative breast cancer (TNBC) subtypes were evaluated by qPCR for the expression of TAM, RON, EPHA2, and MET genes. Statistical analysis was performed to determine the correlation to clinical data. TYRO3 is related to tumor subtype and stage, patient's age, smoking habits, and obesity. MET expression is correlated to EPHA2 and TAM gene expression. EPHA2 expression is also related to aging and smoking habits. The expression levels of the TAM and EPHA2 genes seem to play an important role in breast cancer, being also influenced by the patient's lifestyle.
Collapse
Affiliation(s)
- Ananda Cristina Fernandes de Aguiar
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
| | | | | | | | - Glauber Moreira Leitão
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Clinical Hospital of Pernambuco-Professor Romero Marques, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Luiz Alberto Mattos
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Clinical Hospital of Pernambuco-Professor Romero Marques, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - José Luiz de Lima Filho
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Department of Biochemistry, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| | - Danyelly Bruneska Gondim Martins
- Keizo Asami Institute (iLIKA), Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
- Molecular Prospection and Bioinformatics Group, Recife, Pernambuco, Brazil
- Department of Biochemistry, Federal University of Pernambuco (UFPE), Recife, Pernambuco, Brazil
| |
Collapse
|
6
|
Apostolo D, D’Onghia D, Nerviani A, Ghirardi GM, Sola D, Perazzi M, Tonello S, Colangelo D, Sainaghi PP, Bellan M. Could Gas6/TAM Axis Provide Valuable Insights into the Pathogenesis of Systemic Sclerosis? Curr Issues Mol Biol 2024; 46:7486-7504. [PMID: 39057085 PMCID: PMC11275301 DOI: 10.3390/cimb46070444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disorder characterized by microvascular injury, extracellular matrix deposition, autoimmunity, inflammation, and fibrosis. The clinical complexity and high heterogeneity of the disease make the discovery of potential therapeutic targets difficult. However, the recent progress in the comprehension of its pathogenesis is encouraging. Growth Arrest-Specific 6 (Gas6) and Tyro3, Axl, and MerTK (TAM) receptors are involved in multiple biological processes, including modulation of the immune response, phagocytosis, apoptosis, fibrosis, inflammation, cancer development, and autoimmune disorders. In the present manuscript, we review the current evidence regarding SSc pathogenesis and the role of the Gas6/TAM system in several human diseases, suggesting its likely contribution in SSc and highlighting areas where further research is necessary to fully comprehend the role of TAM receptors in this condition. Indeed, understanding the involvement of TAM receptors in SSc, which is currently unknown, could provide valuable insights for novel potential therapeutic targets.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Davide D’Onghia
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
| | - Alessandra Nerviani
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Giulia Maria Ghirardi
- Centre for Experimental Medicine and Rheumatology, Barts and The London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK;
| | - Daniele Sola
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- IRCCS Istituto Auxologico Italiano, UO General Medicine, 28824 Oggebbio, Italy
| | - Mattia Perazzi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
| | - Stelvio Tonello
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
| | - Donato Colangelo
- Department of Health Sciences, Pharmacology, University of Piemonte Orientale (UPO), 28100 Novara, Italy;
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
- Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (D.D.); (D.S.); (M.P.); (S.T.); (P.P.S.); (M.B.)
- Internal Medicine and Rheumatology Unit, A.O.U. Maggiore della Carità, 28100 Novara, Italy
- Center on Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale, 28100 Novara, Italy
| |
Collapse
|
7
|
Perez F, Iribarren ML, Olexen CM, Ruera CN, Errasti AE, Guzman L, Garbi L, Carrera Silva EA, Chirdo FG. Duodenal mucosa of untreated celiac disease patients has altered expression of the GAS6 and PROS1 and the negative regulator tyrosine kinase TAM receptors subfamily. Clin Immunol 2024; 263:110202. [PMID: 38575045 DOI: 10.1016/j.clim.2024.110202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/14/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.
Collapse
Affiliation(s)
- Federico Perez
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María Luz Iribarren
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Cinthia Mariel Olexen
- Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina (ANM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) de Argentina, Buenos Aires, Argentina
| | - Carolina Naymé Ruera
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Andrea Emilse Errasti
- Instituto de Farmacología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Luciana Guzman
- Servicio de gastroenterología del Hospital de Niños Sor María Ludovica de La Plata, Argentina
| | - Laura Garbi
- Servicio de gastroenterología del Hospital San Martín de la Plata, Argentina
| | - Eugenio Antonio Carrera Silva
- Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina (ANM), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) de Argentina, Buenos Aires, Argentina.
| | - Fernando Gabriel Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), UNLP, CONICET, CIC PBA, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina.
| |
Collapse
|
8
|
Guimarães-Pinto K, Leandro M, Corrêa A, Maia EP, Rodrigues L, da Costa ALA, Rafael Machado Ferreira J, Claudio-Etienne E, Siebenlist U, He J, Rigoni TDS, Ferreira TPT, Jannini-Sa YAP, Matos-Guedes HL, Costa-da-Silva AC, Lopes MF, Silva PMR, Kelsall BL, Filardy AA. Differential regulation of lung homeostasis and silicosis by the TAM receptors MerTk and Axl. Front Immunol 2024; 15:1380628. [PMID: 38774866 PMCID: PMC11106457 DOI: 10.3389/fimmu.2024.1380628] [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: 02/01/2024] [Accepted: 04/10/2024] [Indexed: 05/24/2024] Open
Abstract
Introduction TAM receptor-mediated efferocytosis plays an important function in immune regulation and may contribute to antigen tolerance in the lungs, a site with continuous cellular turnover and generation of apoptotic cells. Some studies have identified failures in efferocytosis as a common driver of inflammation and tissue destruction in lung diseases. Our study is the first to characterize the in vivo function of the TAM receptors, Axl and MerTk, in the innate immune cell compartment, cytokine and chemokine production, as well as the alveolar macrophage (AM) phenotype in different settings in the airways and lung parenchyma. Methods We employed MerTk and Axl defective mice to induce acute silicosis by a single exposure to crystalline silica particles (20 mg/50 μL). Although both mRNA levels of Axl and MerTk receptors were constitutively expressed by lung cells and isolated AMs, we found that MerTk was critical for maintaining lung homeostasis, whereas Axl played a role in the regulation of silica-induced inflammation. Our findings imply that MerTk and Axl differently modulated inflammatory tone via AM and neutrophil recruitment, phenotype and function by flow cytometry, and TGF-β and CXCL1 protein levels, respectively. Finally, Axl expression was upregulated in both MerTk-/- and WT AMs, confirming its importance during inflammation. Conclusion This study provides strong evidence that MerTk and Axl are specialized to orchestrate apoptotic cell clearance across different circumstances and may have important implications for the understanding of pulmonary inflammatory disorders as well as for the development of new approaches to therapy.
Collapse
Affiliation(s)
- Kamila Guimarães-Pinto
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Monique Leandro
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonia Corrêa
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Ester P. Maia
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Leticia Rodrigues
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - André Luiz Amorim da Costa
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Estefannia Claudio-Etienne
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Ulrich Siebenlist
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Jianping He
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Thaís da Silva Rigoni
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | | | - Herbert Leonel Matos-Guedes
- Laboratório de Imunobiotecnologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Laboratório de Imunologia Clínica, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Ana Caroline Costa-da-Silva
- Oral Immunobiology Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health (NIH), Maryland, MD, United States
| | - Marcela Freitas Lopes
- Institute of Biophysics Carlos Chagas Filho, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Brian Lee Kelsall
- Mucosal Immunobiology Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Maryland, MD, United States
| | - Alessandra Almeida Filardy
- Institute of Microbiology, Center for Health Science, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
9
|
Swahn H, Mertens J, Olmer M, Myers K, Mondala TS, Natarajan P, Head SR, Alvarez‐Garcia O, Lotz MK. Shared and Compartment-Specific Processes in Nucleus Pulposus and Annulus Fibrosus During Intervertebral Disc Degeneration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309032. [PMID: 38403470 PMCID: PMC11077672 DOI: 10.1002/advs.202309032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/08/2024] [Indexed: 02/27/2024]
Abstract
Elucidating how cell populations promote onset and progression of intervertebral disc degeneration (IDD) has the potential to enable more precise therapeutic targeting of cells and mechanisms. Single-cell RNA-sequencing (scRNA-seq) is performed on surgically separated annulus fibrosus (AF) (19,978; 26,983 cells) and nucleus pulposus (NP) (20,884; 24,489 cells) from healthy and diseased human intervertebral discs (IVD). In both tissue types, depletion of cell subsets involved in maintenance of healthy IVD is observed, specifically the immature cell subsets - fibroblast progenitors and stem cells - indicative of an impairment of normal tissue self-renewal. Tissue-specific changes are also identified. In NP, several fibrotic populations are increased in degenerated IVD, indicating tissue-remodeling. In degenerated AF, a novel disease-associated subset is identified, which expresses disease-promoting genes. It is associated with pathogenic biological processes and the main gene regulatory networks include thrombospondin signaling and FOXO1 transcription factor. In NP and AF cells thrombospondin protein promoted expression of genes associated with TGFβ/fibrosis signaling, angiogenesis, and nervous system development. The data reveal new insights of both shared and tissue-specific changes in specific cell populations in AF and NP during IVD degeneration. These identified mechanisms and molecules are novel and more precise targets for IDD prevention and treatment.
Collapse
Affiliation(s)
- Hannah Swahn
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| | - Jasmin Mertens
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| | - Merissa Olmer
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| | - Kevin Myers
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| | - Tony S. Mondala
- Center for Computational Biology & Bioinformatics and Genomics CoreScripps ResearchLa JollaCA92037USA
| | - Padmaja Natarajan
- Center for Computational Biology & Bioinformatics and Genomics CoreScripps ResearchLa JollaCA92037USA
| | - Steven R. Head
- Center for Computational Biology & Bioinformatics and Genomics CoreScripps ResearchLa JollaCA92037USA
| | - Oscar Alvarez‐Garcia
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| | - Martin K. Lotz
- Department of Molecular and Cellular Biology & Department of Molecular MedicineScripps ResearchLa JollaCA92037USA
| |
Collapse
|
10
|
Lee YJ, Kim M, Kim HS, Kang JL. Administration of Gas6 attenuates lung fibrosis via inhibition of the epithelial-mesenchymal transition and fibroblast activation. Cell Biol Toxicol 2024; 40:20. [PMID: 38578518 PMCID: PMC10997547 DOI: 10.1007/s10565-024-09858-5] [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: 12/01/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
The epithelial-mesenchymal transition (EMT) and fibroblast activation are major events in idiopathic pulmonary fibrosis pathogenesis. Here, we investigated whether growth arrest-specific protein 6 (Gas6) plays a protective role in lung fibrosis via suppression of the EMT and fibroblast activation. rGas6 administration inhibited the EMT in isolated mouse ATII cells 14 days post-BLM treatment based on morphologic cellular alterations, changes in mRNA and protein expression profiles of EMT markers, and induction of EMT-activating transcription factors. BLM-induced increases in gene expression of fibroblast activation-related markers and the invasive capacity of primary lung fibroblasts in primary lung fibroblasts were reversed by rGas6 administration. Furthermore, the hydroxyproline content and collagen accumulation in interstitial areas with damaged alveolar structures in lung tissue were reduced by rGas6 administration. Targeting Gas6/Axl signaling events with specific inhibitors of Axl (BGB324), COX-2 (NS-398), EP1/EP2 receptor (AH-6809), or PGD2 DP2 receptor (BAY-u3405) reversed the inhibitory effects of rGas6 on EMT and fibroblast activation. Finally, we confirmed the antifibrotic effects of Gas6 using Gas6-/- mice. Therefore, Gas6/Axl signaling events play a potential role in inhibition of EMT process and fibroblast activation via COX-2-derived PGE2 and PGD2 production, ultimately preventing the development of pulmonary fibrosis.
Collapse
Affiliation(s)
- Ye-Ji Lee
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea.
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea.
| |
Collapse
|
11
|
Pan Z, El Sharkway R, Bayoumi A, Metwally M, Gloss BS, Brink R, Lu DB, Liddle C, Alqahtani SA, Yu J, O'Connell PJ, George J, Eslam M. Inhibition of MERTK reduces organ fibrosis in mouse models of fibrotic disease. Sci Transl Med 2024; 16:eadj0133. [PMID: 38569018 DOI: 10.1126/scitranslmed.adj0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
Transforming growth factor-β (TGFβ) drives fibrosis and disease progression in a number of chronic disorders, but targeting this ubiquitously expressed cytokine may not yield a viable and safe antifibrotic therapy. Here, we sought to identify alternative ways to inhibit TGFβ signaling using human hepatic stellate cells and macrophages from humans and mice in vitro, as well as mouse models of liver, kidney, and lung fibrosis. We identified Mer tyrosine kinase (MERTK) as a TGFβ-inducible effector of fibrosis that was up-regulated during fibrosis in multiple organs in three mouse models. We confirmed these findings in liver biopsy samples from patients with metabolic dysfunction-associated fatty liver disease (MAFLD). MERTK also induced TGFβ expression and drove TGFβ signaling resulting in a positive feedback loop that promoted fibrosis in cultured cells. MERTK regulated both canonical and noncanonical TGFβ signaling in both mouse and human cells in vitro. MERTK increased transcription of genes regulating fibrosis by modulating chromatin accessibility and RNA polymerase II activity. In each of the three mouse models, disrupting the fibrosis-promoting signaling loop by reducing MERTK expression reduced organ fibrosis. Pharmacological inhibition of MERTK reduced fibrosis in these mouse models either when initiated immediately after injury or when initiated after fibrosis was established. Together, these data suggest that MERTK plays a role in modulating organ fibrosis and may be a potential target for treating fibrotic diseases.
Collapse
Affiliation(s)
- Ziyan Pan
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Rasha El Sharkway
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Ali Bayoumi
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Mayada Metwally
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Brian S Gloss
- Westmead Research Hub, Westmead Institute for Medical Research, Sydney, NSW 2145, Australia
| | - Robert Brink
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
- St. Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - David Bo Lu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Saleh A Alqahtani
- Organ Transplant Centre of Excellence, King Faisal Specialist Hospital and Research Center, Riyadh 11564, Saudi Arabia
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK-Shenzhen Research Institute, Chinese University of Hong Kong, Hong Kong SAR, PR China
| | - Philip J O'Connell
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW 2145, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, NSW 2145, Australia
| |
Collapse
|
12
|
Miao YR, Rankin EB, Giaccia AJ. Therapeutic targeting of the functionally elusive TAM receptor family. Nat Rev Drug Discov 2024; 23:201-217. [PMID: 38092952 PMCID: PMC11335090 DOI: 10.1038/s41573-023-00846-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 03/07/2024]
Abstract
The TAM receptor family of TYRO3, AXL and MERTK regulates tissue and immune homeostasis. Aberrant TAM receptor signalling has been linked to a range of diseases, including cancer, fibrosis and viral infections. Specifically, the dysregulation of TAM receptors can enhance tumour growth and metastasis due to their involvement in multiple oncogenic pathways. For example, TAM receptors have been implicated in the epithelial-mesenchymal transition, maintaining the stem cell phenotype, immune modulation, proliferation, angiogenesis and resistance to conventional and targeted therapies. Therapeutically, multiple TAM receptor inhibitors are in preclinical and clinical development for cancers and other indications, with those targeting AXL being the most clinically advanced. Although there has been notable clinical advancement in recent years, challenges persist. This Review aims to provide both biological and clinical insights into the current therapeutic landscape of TAM receptor inhibitors, and evaluates their potential for the treatment of cancer and non-malignant diseases.
Collapse
Affiliation(s)
- Yu Rebecca Miao
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erinn B Rankin
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
13
|
Lv H, Qian X, Tao Z, Shu J, Shi D, Yu J, Fan G, Qian Q, Shen L, Lu B. HOXA5-induced lncRNA DNM3OS promotes human embryo lung fibroblast fibrosis via recruiting EZH2 to epigenetically suppress TSC2 expression. J Thorac Dis 2024; 16:1234-1246. [PMID: 38505042 PMCID: PMC10944743 DOI: 10.21037/jtd-23-1145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/01/2023] [Indexed: 03/21/2024]
Abstract
Background Idiopathic pulmonary fibrosis (IPF) is an unrepairable disease that results in lung dysfunction and decreased quality of life. Prevention of pulmonary fibrosis is challenging, while its pathogenesis remains largely unknown. Herein, we investigated the effect and mechanism of long non-coding RNA (lncRNA) DNM3OS/Antisense RNA in the pathogenesis of pulmonary fibrosis. Methods EdU (5-ethynyl-2'-deoxyuridine) and wound healing assays were employed to evaluate the role of DNM3OS on cell proliferation and migration. Western blot detected the proteins expressions of alpha-smooth muscle actin (α-SMA), vimentin, and fibronectin. The interactions among genes were evaluated by RNA pull-down, luciferase reporter, RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP) and chromatin Isolation by RNA purification (ChIRP) assays. Results DNM3OS was upregulated by transforming growth factor beta 1 (TGF-β1) in a dose- and time-dependent manner. DNM3OS knockdown repressed the growth and migration of lung fibroblast, and fibrotic gene expression (CoL1α1, CoL3α1, α-SMA, vimentin, and fibronectin), while suppression of TSC2 accelerated the above process. DNM3OS recruited EZH2 to the promoter region of TSC2, increased the occupancy of EZH2 and H3K27me3, and thereby suppressed the expression of TSC2. HOXA5 promoted the transcription of DNM3OS. Conclusions HOXA5-induced DNM3OS promoted the proliferation, migration, and expression of fibrosis-related genes in human embryo lung fibroblast via recruiting EZH2 to epigenetically suppress the expression of TSC2.
Collapse
Affiliation(s)
- Hong Lv
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Xingjia Qian
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Zhengzheng Tao
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Jun Shu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Dongfang Shi
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Jing Yu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Guiqin Fan
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Qiuhong Qian
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Luhong Shen
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| | - Bing Lu
- Department of Pulmonary and Critical Care Medicine, Taicang TCM Hospital, Affiliated to Nanjing University of Chinese Medicine, Taicang, China
| |
Collapse
|
14
|
Tutusaus A, Morales A, García de Frutos P, Marí M. GAS6/TAM Axis as Therapeutic Target in Liver Diseases. Semin Liver Dis 2024; 44:99-114. [PMID: 38395061 PMCID: PMC11027478 DOI: 10.1055/a-2275-0408] [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] [Indexed: 02/25/2024]
Abstract
TAM (TYRO3, AXL, and MERTK) protein tyrosine kinase membrane receptors and their vitamin K-dependent ligands GAS6 and protein S (PROS) are well-known players in tumor biology and autoimmune diseases. In contrast, TAM regulation of fibrogenesis and the inflammation mechanisms underlying metabolic dysfunction-associated steatohepatitis (MASH), cirrhosis, and, ultimately, liver cancer has recently been revealed. GAS6 and PROS binding to phosphatidylserine exposed in outer membranes of apoptotic cells links TAMs, particularly MERTK, with hepatocellular damage. In addition, AXL and MERTK regulate the development of liver fibrosis and inflammation in chronic liver diseases. Acute hepatic injury is also mediated by the TAM system, as recent data regarding acetaminophen toxicity and acute-on-chronic liver failure have uncovered. Soluble TAM-related proteins, mainly released from activated macrophages and hepatic stellate cells after hepatic deterioration, are proposed as early serum markers for disease progression. In conclusion, the TAM system is becoming an interesting pharmacological target in liver pathology and a focus of future biomedical research in this field.
Collapse
Affiliation(s)
- Anna Tutusaus
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| | - Pablo García de Frutos
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Cardiovasculares (CIBERCV), Barcelona, Comunidad de Madrid, Spain
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Barcelona, Catalunya, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, Barcelona, Spain
| |
Collapse
|
15
|
Espindola MS, Habiel DM, Coelho AL, Parimon T, Chen P, Mikels-Vigdal A, Hogaboam CM. Translational Studies Reveal the Divergent Effects of Simtuzumab Targeting LOXL2 in Idiopathic Pulmonary Fibrosis. FIBROSIS (HONG KONG, CHINA) 2023; 1:10007. [PMID: 38873180 PMCID: PMC11175361 DOI: 10.35534/fibrosis.2023.10007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The composition of extracellular matrix (ECM) is altered during pathologic scarring in damaged organs including the lung. One major change in the ECM involves the cross-linking of collagen, which promotes fibroblast to myofibroblast differentiation. We examined the role of lysyl oxidase (LOX)-like 2 in lung progenitors and fibroblasts cultured from normal or IPF lung samples and in a humanized mouse model of IPF using a monoclonal antibody (Simtuzumab). Primary lung fibroblasts from normal donor lungs and IPF lung explants were examined for expression of LOXL2. Targeting LOXL2 with Simtuzumab on normal and IPF fibroblasts was examined both in vitro and in vivo for synthetic, functional, and profibrotic properties. LOXL2 was increased at transcript and protein level in IPF compared with normal lung samples. In a dose-dependent manner, Simtuzumab enhanced differentiation of fibroblasts into myofibroblasts. Inhibition of LOXL2 also enhanced fibroblast invasion and accelerated the outgrowth of fibroblasts from dissociated human lung cell preparations. Finally, preventative or delayed delivery of Simtuzumab enhanced lung fibrosis in a humanized mouse model of pulmonary fibrosis. Consistent with its failure in a Phase 2 clinical trial, Simtuzumab exhibited no therapeutic efficacy in translational in vitro and in vivo assays.
Collapse
Affiliation(s)
- Milena S. Espindola
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David M. Habiel
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ana Lucia Coelho
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tanyalak Parimon
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Peter Chen
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - Cory M. Hogaboam
- Women’s Guild Lung Institute, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
16
|
Wang L, Li Z, Wan R, Pan X, Li B, Zhao H, Yang J, Zhao W, Wang S, Wang Q, Yan P, Ma C, Yuan H, Zhao M, Rosas I, Ding C, Sun B, Yu G. Single-Cell RNA Sequencing Provides New Insights into Therapeutic Roles of Thyroid Hormone in Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2023; 69:456-469. [PMID: 37402274 PMCID: PMC10557923 DOI: 10.1165/rcmb.2023-0080oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/29/2023] [Indexed: 07/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fatal interstitial lung disease without an effective cure. Herein, we explore the role of 3,5,3'-triiodothyronine (T3) administration on lung alveolar regeneration and fibrosis at the single-cell level. T3 supplementation significantly altered the gene expression in fibrotic lung tissues. Immune cells were rapidly recruited into the lung after the injury; there were much more M2 macrophages than M1 macrophages in the lungs of bleomycin-treated mice; and M1 macrophages increased slightly, whereas M2 macrophages were significantly reduced after T3 treatment. T3 enhanced the resolution of pulmonary fibrosis by promoting the differentiation of Krt8+ transitional alveolar type II epithelial cells into alveolar type I epithelial cells and inhibiting fibroblast activation and extracellular matrix production potentially by regulation of Nr2f2. In addition, T3 regulated the crosstalk of macrophages with fibroblasts, and the Pros1-Axl signaling axis significantly facilitated the attenuation of fibrosis. The findings demonstrate that administration of a thyroid hormone promotes alveolar regeneration and resolves fibrosis mainly by regulation of the cellular state and cell-cell communication of alveolar epithelial cells, macrophages, and fibroblasts in mouse lungs in comprehensive ways.
Collapse
Affiliation(s)
- Lan Wang
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Zhongzheng Li
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Ruyan Wan
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Xin Pan
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Bin Li
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Huabin Zhao
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Juntang Yang
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Weiming Zhao
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Shenghui Wang
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Qiwen Wang
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Peishuo Yan
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Chi Ma
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
- School of Life Sciences, Fudan University, Shanghai, China; and
| | - Hongmei Yuan
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Mengxia Zhao
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Ivan Rosas
- Division of Pulmonary, Critical Care and Sleep Medicine, Baylor College of Medicine, Houston, Texas
| | - Chen Ding
- School of Life Sciences, Fudan University, Shanghai, China; and
| | - Baofa Sun
- College of Life Science, Nankai University, Tianjin, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, and
- Henan International Joint Laboratory of Pulmonary Fibrosis, College of Life Science, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
17
|
Nordbø OP, Landolt L, Eikrem Ø, Scherer A, Leh S, Furriol J, Apeland T, Mydel P, Marti H. Transcriptomic analysis reveals partial epithelial-mesenchymal transition and inflammation as common pathogenic mechanisms in hypertensive nephrosclerosis and Type 2 diabetic nephropathy. Physiol Rep 2023; 11:e15825. [PMID: 37813528 PMCID: PMC10562137 DOI: 10.14814/phy2.15825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 10/12/2023] Open
Abstract
Hypertensive nephrosclerosis (HN) and Type 2 diabetic nephropathy (T2DN) are the leading causes of chronic kidney disease (CKD). To explore shared pathogenetic mechanisms, we analyzed transcriptomes of kidney biopsies from patients with HN or T2DN. Total RNA was extracted from 10 μm whole kidney sections from patients with HN, T2DN, and normal controls (Ctrl) (n = 6 for each group) and processed for RNA sequencing. Differentially expressed (log2 fold change >1, adjusted p < 0.05) genes (DEG) and molecular pathways were analyzed, and selected results were validated by immunohistochemistry (IHC). ELISA on serum samples was performed on a related cohort consisting of patients with biopsy-proven HN (n = 13) and DN (n = 9), and a normal control group (n = 14). Cluster analysis on RNA sequencing data separated diseased and normal tissues. RNA sequencing revealed that 88% (341 out of 384) of DEG in HN were also altered in T2DN, while gene set enrichment analysis (GSEA) showed that over 90% of affected molecular pathways, including those related to inflammation, immune response, and cell-cycle regulation, were similarly impacted in both HN and T2DN samples. The increased expression of genes tied to interleukin signaling and lymphocyte activation was more pronounced in HN, while genes associated with extracellular matrix organization were more evident in T2DN. Both HN and T2DN tissues exhibited significant upregulation of genes connected with inflammatory responses, T-cell activity, and partial epithelial to mesenchymal transition (p-EMT). Immunohistochemistry (IHC) further confirmed T-cell (CD4+ and CD8+ ) infiltration in the diseased tissues. Additionally, IHC revealed heightened AXL protein expression, a key regulator of inflammation and p-EMT, in both HN and T2DN, while serum analysis indicated elevated soluble AXL levels in patients with both conditions. These findings underline the shared molecular mechanisms between HN and T2DN, hinting at the potential for common therapeutic strategies targeting both diseases.
Collapse
Affiliation(s)
- Ole Petter Nordbø
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of Medicine, Haugesund HospitalHelse FonnaHaugesundNorway
| | - Lea Landolt
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| | - Øystein Eikrem
- Department of Clinical ScienceUniversity of BergenBergenNorway
| | | | - Sabine Leh
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of PathologyHaukeland University HospitalBergenNorway
| | - Jessica Furriol
- Department of Clinical MedicineUniversity of BergenBergenNorway
| | | | - Piotr Mydel
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| | - Hans‐Peter Marti
- Department of Clinical MedicineUniversity of BergenBergenNorway
- Department of MedicineHaukeland University HospitalBergenNorway
| |
Collapse
|
18
|
Hewitt RJ, Puttur F, Gaboriau DCA, Fercoq F, Fresquet M, Traves WJ, Yates LL, Walker SA, Molyneaux PL, Kemp SV, Nicholson AG, Rice A, Roberts E, Lennon R, Carlin LM, Byrne AJ, Maher TM, Lloyd CM. Lung extracellular matrix modulates KRT5 + basal cell activity in pulmonary fibrosis. Nat Commun 2023; 14:6039. [PMID: 37758700 PMCID: PMC10533905 DOI: 10.1038/s41467-023-41621-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Aberrant expansion of KRT5+ basal cells in the distal lung accompanies progressive alveolar epithelial cell loss and tissue remodelling during fibrogenesis in idiopathic pulmonary fibrosis (IPF). The mechanisms determining activity of KRT5+ cells in IPF have not been delineated. Here, we reveal a potential mechanism by which KRT5+ cells migrate within the fibrotic lung, navigating regional differences in collagen topography. In vitro, KRT5+ cell migratory characteristics and expression of remodelling genes are modulated by extracellular matrix (ECM) composition and organisation. Mass spectrometry- based proteomics revealed compositional differences in ECM components secreted by primary human lung fibroblasts (HLF) from IPF patients compared to controls. Over-expression of ECM glycoprotein, Secreted Protein Acidic and Cysteine Rich (SPARC) in the IPF HLF matrix restricts KRT5+ cell migration in vitro. Together, our findings demonstrate how changes to the ECM in IPF directly influence KRT5+ cell behaviour and function contributing to remodelling events in the fibrotic niche.
Collapse
Affiliation(s)
- Richard J Hewitt
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Franz Puttur
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - David C A Gaboriau
- Facility for Imaging by Light Microscopy, National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | | | - Maryline Fresquet
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - William J Traves
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Laura L Yates
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Simone A Walker
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Philip L Molyneaux
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Samuel V Kemp
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
- Department of Respiratory Medicine, Nottingham University Hospitals NHS Trust, City Campus, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Andrew G Nicholson
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Alexandra Rice
- Royal Brompton and Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, SW3 6NP, UK
| | - Edward Roberts
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Leo M Carlin
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Adam J Byrne
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
| | - Toby M Maher
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Keck Medicine of USC, 1510 San Pablo Street, Los Angeles, CA, 90033, USA
| | - Clare M Lloyd
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
19
|
Yu S, Kalinin AA, Paraskevopoulou MD, Maruggi M, Cheng J, Tang J, Icke I, Luo Y, Wei Q, Scheibe D, Hunter J, Singh S, Nguyen D, Carpenter AE, Horman SR. Integrating inflammatory biomarker analysis and artificial-intelligence-enabled image-based profiling to identify drug targets for intestinal fibrosis. Cell Chem Biol 2023; 30:1169-1182.e8. [PMID: 37437569 PMCID: PMC10529501 DOI: 10.1016/j.chembiol.2023.06.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 03/11/2023] [Accepted: 06/13/2023] [Indexed: 07/14/2023]
Abstract
Intestinal fibrosis, often caused by inflammatory bowel disease, can lead to intestinal stenosis and obstruction, but there are no approved treatments. Drug discovery has been hindered by the lack of screenable cellular phenotypes. To address this, we used a scalable image-based morphology assay called Cell Painting, augmented with machine learning algorithms, to identify small molecules that could reverse the activated fibrotic phenotype of intestinal myofibroblasts. We then conducted a high-throughput small molecule chemogenomics screen of approximately 5,000 compounds with known targets or mechanisms, which have achieved clinical stage or approval by the FDA. By integrating morphological analyses and AI using pathologically relevant cells and disease-relevant stimuli, we identified several compounds and target classes that are potentially able to treat intestinal fibrosis. This phenotypic screening platform offers significant improvements over conventional methods for identifying a wide range of drug targets.
Collapse
Affiliation(s)
- Shan Yu
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| | | | | | - Marco Maruggi
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Jie Cheng
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Jie Tang
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Ilknur Icke
- Takeda Development Center Americas, Inc., Cambridge, MA 02142, USA
| | - Yi Luo
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Qun Wei
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Dan Scheibe
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Joel Hunter
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | - Shantanu Singh
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Deborah Nguyen
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA
| | | | - Shane R Horman
- Takeda Development Center Americas, Inc., San Diego, CA 92121, USA.
| |
Collapse
|
20
|
Apostolo D, Ferreira LL, Di Tizio A, Ruaro B, Patrucco F, Bellan M. A Review: The Potential Involvement of Growth Arrest-Specific 6 and Its Receptors in the Pathogenesis of Lung Damage and in Coronavirus Disease 2019. Microorganisms 2023; 11:2038. [PMID: 37630598 PMCID: PMC10459962 DOI: 10.3390/microorganisms11082038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
The tyrosine kinase receptors of the TAM family-Tyro3, Axl and Mer-and their main ligand Gas6 (growth arrest-specific 6) have been implicated in several human diseases, having a particularly important role in the regulation of innate immunity and inflammatory response. The Gas6/TAM system is involved in the recognition of apoptotic debris by immune cells and this mechanism has been exploited by viruses for cell entry and infection. Coronavirus disease 2019 (COVID-19) is a multi-systemic disease, but the lungs are particularly affected during the acute phase and some patients may suffer persistent lung damage. Among the manifestations of the disease, fibrotic abnormalities have been observed among the survivors of COVID-19. The mechanisms of COVID-related fibrosis remain elusive, even though some parallels may be drawn with other fibrotic diseases, such as idiopathic pulmonary fibrosis. Due to the still limited number of scientific studies addressing this question, in this review we aimed to integrate the current knowledge of the Gas6/TAM axis with the pathophysiological mechanisms underlying COVID-19, with emphasis on the development of a fibrotic phenotype.
Collapse
Affiliation(s)
- Daria Apostolo
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
| | - Luciana L. Ferreira
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
| | - Alice Di Tizio
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
- Respiratory Diseases Unit, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Barbara Ruaro
- Pulmonology Department, University of Trieste, 34128 Trieste, Italy;
| | - Filippo Patrucco
- Respiratory Diseases Unit, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, University of Piemonte Orientale (UPO), 28100 Novara, Italy; (D.A.); (L.L.F.); (A.D.T.); (M.B.)
- Division of Internal Medicine, Medical Department, AOU Maggiore della Carità Hospital, 28100 Novara, Italy
| |
Collapse
|
21
|
Yong J, Elisabeth Groeger S, Ruf S, Ruiz-Heiland G. Influence of leptin and compression in GAS-6 mediated homeostasis of periodontal ligament cell. Oral Dis 2023; 29:1172-1183. [PMID: 34861742 DOI: 10.1111/odi.14092] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 11/21/2021] [Accepted: 12/01/2021] [Indexed: 11/29/2022]
Abstract
Growth arrest-specific protein 6 (GAS-6) regulates immunomodulatory and inflammatory mechanisms in periodontium and may participate in obesity predisposition. This study aimed to determine whether GAS-6 is associated with the homeostasis of periodontal ligament (SV-PDL) cells in the presence of adipokines or compressive forces. The SV-PDL cell line was used. Western blots were employed for TAM receptors detection. Cells were stimulated using different concentrations of GAS-6. The migration, viability, and proliferation were measured by a standard scratch test, MTS assay, and immunofluorescent staining. The mRNA expression was analyzed by RT-PCR. Release of TGF-β1, GAS-6, and Axl were verified by ELISA. Western blot shows that TAM receptors are expressed in SV-PDL cells. GAS-6 has a promoting effect on cell migration and proliferation. RT-PCR analysis showed that GAS-6 induces Collagen-1, Collagen-3, Periostin, and TGF-β1 mRNA expression whereas it reduces Caspase-3, Caspase-8, Caspase-9, and IL-6 mRNA expression. Further, secreted GAS-6 in SV-PDL is reduced in response to both compressive forces and leptin and upregulated by IL-6. Additionally, ADAM-10 inhibition reduces GAS-6 and Axl release on SV-PDL cells. TAM receptors especially Axl are identified as the receptors of GAS-6. GAS-6/TAM interactions contribute to periodontal ligament cells homeostasis. Leptin inhibits the GAS-6 release independently of ADAM-10 metalloprotease.
Collapse
Affiliation(s)
- Jiawen Yong
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Elisabeth Groeger
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Sabine Ruf
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Gisela Ruiz-Heiland
- Department of Orthodontics, Faculty of Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
22
|
Liu HJ, Miao H, Yang JZ, Liu F, Cao G, Zhao YY. Deciphering the role of lipoproteins and lipid metabolic alterations in ageing and ageing-associated renal fibrosis. Ageing Res Rev 2023; 85:101861. [PMID: 36693450 DOI: 10.1016/j.arr.2023.101861] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/07/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Fibrosis is the ultimate pathological feature of many chronic diseases, and ageing a major risk factor for fibrotic diseases. Current therapies are limited to those that reduce the rate of functional decline in patients with mild to moderate disease, but few interventions are available to specifically target the pathogenesis of fibrosis. In this context, new treatments that can significantly improve survival time and quality of life for these patients are urgently needed. In this review, we outline both the synthesis and metabolism of lipids and lipoproteins associated with ageing-associated renal fibrosis and the prominent contribution of lipids and lipidomics in the discovery of biomarkers that can be used for the prevention, diagnosis, and treatment of renal ageing and fibrosis. Next, we describe the effect of dyslipidaemia on ageing-related renal fibrosis and the pathophysiological changes in the kidney caused by dyslipidaemia. We then summarize the enzymes, transporters, transcription factors, and RNAs that contribute to dysregulated lipid metabolism in renal fibrosis and discuss their role in renal fibrosis in detail. We conclude by discussing the progress in research on small molecule therapeutic agents that prevent and treat ageing and ageing-associated renal fibrosis by modulating lipid metabolism. A growing number of studies suggest that restoring aberrant lipid metabolism may be a novel and promising therapeutic strategy to combat ageing and ageing-associated renal fibrosis.
Collapse
Affiliation(s)
- Hong-Jiao Liu
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China
| | - Jun-Zheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, No. 71 Dongpeng Avenue, Guangzhou, Guangdong 510530, China
| | - Fei Liu
- Department of Urology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17 South of Panjiayuan, Beijing 100021, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China.
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
23
|
Plasma GAS6 predicts mortality risk in acute heart failure patients: insights from the DRAGON-HF trial. J Transl Med 2023; 21:21. [PMID: 36635690 PMCID: PMC9838057 DOI: 10.1186/s12967-022-03859-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Growth arrest-specific 6 (GAS6) is a vitamin K-dependent protein related to inflammation, fibrosis, as well as platelet function. Genetic ablation of GAS6 in mice protects against cardiac hypertrophy and dysfunction. Nonetheless, the association between plasma GAS6 levels and acute heart failure (AHF) patients is still unknown. METHODS We measured plasma GAS6 concentrations in 1039 patients with AHF who were enrolled in the DRAGON-HF trial (NCT03727828). Mean follow-up of the study was 889 days. The primary endpoint is all-cause death. RESULTS In total, there were 195 primary endpoints of all-cause death and 135 secondary endpoints of cardiovascular death during the mean follow-up duration of 889 days. The higher levels of GAS6 were associated with higher rates of all-cause and cardiovascular death (P < 0.05). Baseline plasma GAS6 levels were still strongly correlated with clinical outcomes in different models after adjustment for clinical factors and N-terminal pro-brain natriuretic peptide (NT-proBNP, P < 0.05). GAS6 could further distinguish the risks of clinical outcomes based on NT-proBNP measurement. CONCLUSION Elevated plasma GAS6 levels were associated with an increased risk of all-cause and cardiovascular death in patients with AHF. Trial registration NCT03727828 (DRAGON-HF trial) clinicaltrials.gov.
Collapse
|
24
|
Rizzi M, Tonello S, D’Onghia D, Sainaghi PP. Gas6/TAM Axis Involvement in Modulating Inflammation and Fibrosis in COVID-19 Patients. Int J Mol Sci 2023; 24:ijms24020951. [PMID: 36674471 PMCID: PMC9861142 DOI: 10.3390/ijms24020951] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Gas6 (growth arrest-specific gene 6) is a widely expressed vitamin K-dependent protein that is involved in many biological processes such as homeostatic regulation, inflammation and repair/fibrotic processes. It is known that it is the main ligand of TAMs, a tyrosine kinase receptor family of three members, namely MerTK, Tyro-3 and Axl, for which it displays the highest affinity. Gas6/TAM axis activation is known to be involved in modulating inflammatory responses as well as fibrotic evolution in many different pathological conditions. Due to the rapidly evolving COVID-19 pandemic, this review will focus on Gas6/TAM axis activation in SARS-CoV-2 infection, where de-regulated inflammatory responses and fibrosis represent a relevant feature of severe disease manifestation. Furthermore, this review will highlight the most recent scientific evidence supporting an unsuspected role of Axl as a SARS-CoV-2 infection driver, and the potential therapeutic advantages of the use of existing Axl inhibitors in COVID-19 management. From a physiological point of view, the Gas6/TAM axis plays a dual role, fostering the tissue repair processes or leading to organ damage and loss of function, depending on the prevalence of its anti-inflammatory or profibrotic properties. This review makes a strong case for further research focusing on the Gas6/TAM axis as a pharmacological target to manage different disease conditions, such as chronic fibrosis or COVID-19.
Collapse
|
25
|
He Y, Huang J, Li Q, Xia W, Zhang C, Liu Z, Xiao J, Yi Z, Deng H, Xiao Z, Hu J, Li H, Zu X, Quan C, Chen J. Gut Microbiota and Tumor Immune Escape: A New Perspective for Improving Tumor Immunotherapy. Cancers (Basel) 2022; 14:5317. [PMID: 36358736 PMCID: PMC9656981 DOI: 10.3390/cancers14215317] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 10/15/2023] Open
Abstract
The gut microbiota is a large symbiotic community of anaerobic and facultative aerobic bacteria inhabiting the human intestinal tract, and its activities significantly affect human health. Increasing evidence has suggested that the gut microbiome plays an important role in tumor-related immune regulation. In the tumor microenvironment (TME), the gut microbiome and its metabolites affect the differentiation and function of immune cells regulating the immune evasion of tumors. The gut microbiome can indirectly influence individual responses to various classical tumor immunotherapies, including immune checkpoint inhibitor therapy and adoptive immunotherapy. Microbial regulation through antibiotics, prebiotics, and fecal microbiota transplantation (FMT) optimize the composition of the gut microbiome, improving the efficacy of immunotherapy and bringing a new perspective and hope for tumor treatment.
Collapse
Affiliation(s)
- Yunbo He
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Qiaorong Li
- Department of Ultrasound, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410000, China
| | - Weiping Xia
- Department of Intensive Care Medicine, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Chunyu Zhang
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhi Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Hao Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Zicheng Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Chao Quan
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410013, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410013, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410013, China
| |
Collapse
|
26
|
Xie Y, Ma J, Xie L, Li W, Yang M, Gu P, Zhang Y, Fan L, Wang D, Chen W. Inhibition of Gas6 promotes crystalline silica-induced inflammatory response of macrophages via blocking autophagy flux. ENVIRONMENTAL TOXICOLOGY 2022; 37:1925-1933. [PMID: 35438832 DOI: 10.1002/tox.23539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/29/2022] [Accepted: 04/10/2022] [Indexed: 06/14/2023]
Abstract
Inhalation of crystalline silica (CS) can cause silicosis, which is one of the most serious interstitial lung diseases worldwide. Autophagy dysfunction is an essential step in silicosis progression. In this study, we aim to identify the effect of growth arrest-specific protein 6 (Gas6) during autophagy induction and macrophage inflammatory response caused by CS. After RAW 264.7 macrophages exposed to CS, the levels of Gas6 and autophagy markers (p62, Beclin1, and LC3-II/LC3-I) were increased, accompanied with enhanced inflammatory cytokines secretion. Using autophagy activator (rapamycin) repressed, whereas autophagy inhibitor (3-methyladenine) promoted inflammatory cytokines release. Besides, inhibition of Gas6 aggravated CS-induced inflammatory response, and autophagy inhibition facilitated the promoted effect of Gas6 silencing, resulting in elevated expression of inflammatory cytokines. These findings reveal the protective effects of Gas6 and autophagy in macrophages in response to CS exposure, and highlight the autophagy regulated by Gas6 may be a potential prevention target for CS-induced lung inflammatory response.
Collapse
Affiliation(s)
- Yujia Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jixuan Ma
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Li Xie
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Li
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Meng Yang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Pei Gu
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yingdie Zhang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lieyang Fan
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dongming Wang
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weihong Chen
- Department of Occupational & Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
27
|
Ma H, Liu S, Li S, Xia Y. Targeting Growth Factor and Cytokine Pathways to Treat Idiopathic Pulmonary Fibrosis. Front Pharmacol 2022; 13:918771. [PMID: 35721111 PMCID: PMC9204157 DOI: 10.3389/fphar.2022.918771] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/06/2022] [Indexed: 02/05/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial lung disease of unknown origin that usually results in death from secondary respiratory failure within 2–5 years of diagnosis. Recent studies have identified key roles of cytokine and growth factor pathways in the pathogenesis of IPF. Although there have been numerous clinical trials of drugs investigating their efficacy in the treatment of IPF, only Pirfenidone and Nintedanib have been approved by the FDA. However, they have some major limitations, such as insufficient efficacy, undesired side effects and poor pharmacokinetic properties. To give more insights into the discovery of potential targets for the treatment of IPF, this review provides an overview of cytokines, growth factors and their signaling pathways in IPF, which have important implications for fully exploiting the therapeutic potential of targeting cytokine and growth factor pathways. Advances in the field of cytokine and growth factor pathways will help slow disease progression, prolong life, and improve the quality of life for IPF patients in the future.
Collapse
Affiliation(s)
- Hongbo Ma
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shengming Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Shanrui Li
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| |
Collapse
|
28
|
Baseline Plasma Gas6 Protein Elevation Predicts Adverse Outcomes in Hospitalized COVID-19 Patients. DISEASE MARKERS 2022; 2022:1568352. [PMID: 35531477 PMCID: PMC9070408 DOI: 10.1155/2022/1568352] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/18/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022]
Abstract
Reliable biomarkers allowing early patients’ stratification for the risk of adverse outcomes in COVID-19 are lacking. Gas6, together with its tyrosine kinase receptors named TAM, is involved in the regulation of immune homeostasis, fibrosis, and thrombosis. Our aim was to evaluate whether Gas6, sAxl, and sMerTK could represent early predictors of disease evolution either towards a negative (death or need of ICU admission) or a positive (discharge and/or clinical resolution within the first 14 days of hospitalization) outcome. To this purpose, between January and May 2021 (corresponding to third pandemic wave in Italy), 139 consecutive SARS-CoV-2 positive patients were enrolled in a prospective observational study. Plasma levels of these molecules were measured by ELISA at the time of hospitalization and after 7 and 14 days. We observed that higher plasma Gas6 concentrations at hospital admission were associated with a worsening in clinical conditions while lower sMerTK concentrations at baseline and after 7 days of hospitalization were associated with a more favorable outcome. At multivariate analysis, after correction for demographic and COVID-19 severity variables (NEWS2 and PiO2/FiO2), only Gas6 measured at baseline predicted an adverse prognosis with an odds ratio of 1.03 (C.I. 1.01-10.5). At ROC curve analysis, baseline Gas6 levels higher than 58.0 ng/ml predicted a severe disease evolution with 53.3% sensitivity and 77.6% specificity (area under the curve 0.653,
, likelihood ratio of 2.38, IQR: 1.46-3.87). Taken together, these results support the hypothesis that a dysregulation in the Gas6/TAM axis could play a relevant role in modulating the course of COVID-19 and suggest that plasma Gas6 may represent a promising prognostic laboratory parameter for this condition.
Collapse
|
29
|
Valdoz JC, Franks NA, Cribbs CG, Jacobs DJ, Dodson EL, Knight CJ, Poulson PD, Garfield SR, Johnson BC, Hemeyer BM, Sudo MT, Saunooke JA, Kartchner BC, Saxton A, Vallecillo-Zuniga ML, Santos M, Chamberlain B, Christensen KA, Nordin GP, Narayanan AS, Raghu G, Van Ry PM. Soluble ECM promotes organotypic formation in lung alveolar model. Biomaterials 2022; 283:121464. [DOI: 10.1016/j.biomaterials.2022.121464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/15/2022] [Accepted: 03/06/2022] [Indexed: 11/25/2022]
|
30
|
Zhang J, Liu Q, He J, Li Y. Novel Therapeutic Targets in Liver Fibrosis. Front Mol Biosci 2021; 8:766855. [PMID: 34805276 PMCID: PMC8602792 DOI: 10.3389/fmolb.2021.766855] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/18/2021] [Indexed: 02/05/2023] Open
Abstract
Liver fibrosis is end-stage liver disease that can be rescued. If irritation continues due to viral infection, schistosomiasis and alcoholism, liver fibrosis can progress to liver cirrhosis and even cancer. The US Food and Drug Administration has not approved any drugs that act directly against liver fibrosis. The only treatments currently available are drugs that eliminate pathogenic factors, which show poor efficacy; and liver transplantation, which is expensive. This highlights the importance of clarifying the mechanism of liver fibrosis and searching for new treatments against it. This review summarizes how parenchymal, nonparenchymal cells, inflammatory cells and various processes (liver fibrosis, hepatic stellate cell activation, cell death and proliferation, deposition of extracellular matrix, cell metabolism, inflammation and epigenetics) contribute to liver fibrosis. We highlight discoveries of novel therapeutic targets, which may provide new insights into potential treatments for liver fibrosis.
Collapse
Affiliation(s)
- Jinhang Zhang
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Qinhui Liu
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| | - Jinhan He
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China.,Department of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Sichuan, China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
31
|
Abstract
Extensive interest in cancer immunotherapy is reported according to the clinical importance of CTLA-4 and (PD-1/PD-L1) [programmed death (PD) and programmed death-ligand (PD-L1)] in immune checkpoint therapies. AXL is a receptor tyrosine kinase expressed in different types of cancer and in relation to resistance against various anticancer therapeutics due to poor clinical prognosis. AXL and its ligand, i.e., growth arrest-specific 6 (GAS6) proteins, are expressed on many cancer cells, and the GAS6/AXL pathway is reported to promote cancer cell proliferation, survival, migration, invasion, angiogenesis, and immune evasion. AXL is an attractive and novel therapeutic target for impairing tumor progression from immune cell contracts in the tumor microenvironment. The GAS6/AXL pathway is also of interest immunologically because it targets fewer antitumor immune responses. In effect, several targeted therapies are selective and nonselective for AXL, which are in preclinical and clinical development in multiple cancer types. Therefore, this review focuses on the role of the GAS6/AXL signaling pathway in triggering the immunosuppressive tumor microenvironment as immune evasion. This includes regulating its composition and activating T-cell exclusion with the immune-suppressive activity of regulatory T cells, which is related to one of the hallmarks of cancer survival. Finally, this article discusses the GAS6/AXL signaling pathway in the context of several immune responses such as NK cell activation, apoptosis, and tumor-specific immunity, especially PD-1/PDL-1 signaling.
Collapse
Affiliation(s)
- Hye-Youn Son
- Department of Breast and Endocrine Surgery, Center for Medical Innovation, Seoul National University Hospital, Seoul, South Korea
| | - Hwan-Kyu Jeong
- School of Biosystems and Biomedical Sciences, Korea University, Seoul, South Korea
| |
Collapse
|
32
|
Kim BM, Lee YJ, Choi YH, Park EM, Kang JL. Gas6 Ameliorates Inflammatory Response and Apoptosis in Bleomycin-Induced Acute Lung Injury. Biomedicines 2021; 9:1674. [PMID: 34829903 PMCID: PMC8615678 DOI: 10.3390/biomedicines9111674] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/07/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Acute lung injury (ALI) is characterized by alveolar damage, lung edema, and exacerbated inflammatory response. Growth arrest-specific protein 6 (Gas6) mediates many different functions, including cell survival, proliferation, inflammatory signaling, and apoptotic cell clearance (efferocytosis). The role of Gas6 in bleomycin (BLM)-induced ALI is unknown. We investigated whether exogenous administration of mouse recombinant Gas6 (rGas6) has anti-inflammatory and anti-apoptotic effects on BLM-induced ALI. Compared to mice treated with only BLM, the administration of rGas6 reduced the secretion of proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and macrophage inflammatory protein-2, and increased the secretion of hepatocyte growth factor in bronchoalveolar lavage (BAL) fluid. rGas6 administration also reduced BLM-induced inflammation and apoptosis as evidenced by reduced neutrophil recruitment into the lungs, total protein levels in BAL fluid, caspase-3 activity, and TUNEL-positive lung cells in lung tissue. Apoptotic cell clearance by alveolar macrophages was also enhanced in mice treated with both BLM and rGas6 compared with mice treated with only BLM. rGas6 also had pro-resolving and anti-apoptotic effects in mouse bone marrow-derived macrophages and alveolar epithelial cell lines stimulated with BLM in vitro. These findings indicate that rGas6 may play a protective role in BLM-induced ALI.
Collapse
Affiliation(s)
- Bo-Min Kim
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (B.-M.K.); (Y.-J.L.); (Y.-H.C.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Ye-Ji Lee
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (B.-M.K.); (Y.-J.L.); (Y.-H.C.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Youn-Hee Choi
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (B.-M.K.); (Y.-J.L.); (Y.-H.C.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| | - Eun-Mi Park
- Department of Pharmacology, College of Medicine, Ewha Womans University, Seoul 07804, Korea;
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, Seoul 07804, Korea; (B.-M.K.); (Y.-J.L.); (Y.-H.C.)
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, Seoul 07804, Korea
| |
Collapse
|
33
|
Pradhan RN, Krishnamurty AT, Fletcher AL, Turley SJ, Müller S. A bird's eye view of fibroblast heterogeneity: A pan-disease, pan-cancer perspective. Immunol Rev 2021; 302:299-320. [PMID: 34164824 DOI: 10.1111/imr.12990] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/14/2021] [Indexed: 02/07/2023]
Abstract
Fibroblasts, custodians of tissue architecture and function, are no longer considered a monolithic entity across tissues and disease indications. Recent advances in single-cell technologies provide an unrestricted, high-resolution view of fibroblast heterogeneity that exists within and across tissues. In this review, we summarize a compendium of single-cell transcriptomic studies and provide a comprehensive accounting of fibroblast subsets, many of which have been described to occupy specific niches in tissues at homeostatic and pathologic states. Understanding this heterogeneity is particularly important in the context of cancer, as the diverse cancer-associated fibroblast (CAF) phenotypes in the tumor microenvironment (TME) are directly impacted by the expression phenotypes of their predecessors. Relationships between these heterogeneous populations often accompany and influence response to therapy in cancer and fibrosis. We further highlight the importance of integrating single-cell studies to deduce common fibroblast phenotypes across disease states, which will facilitate the identification of common signaling pathways, gene regulatory programs, and cell surface markers that are going to advance drug discovery and targeting.
Collapse
|
34
|
Yang DC, Gu S, Li JM, Hsu SW, Chen SJ, Chang WH, Chen CH. Targeting the AXL Receptor in Combating Smoking-related Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2021; 64:734-746. [PMID: 33730527 PMCID: PMC8456879 DOI: 10.1165/rcmb.2020-0303oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 02/01/2021] [Indexed: 11/24/2022] Open
Abstract
Tobacco smoking is a well-known risk factor for both fibrogenesis and fibrotic progression; however, the mechanisms behind these processes remain enigmatic. RTKs (receptor tyrosine kinases) have recently been reported to drive profibrotic phenotypes in fibroblasts during pulmonary fibrosis (PF). Using a phospho-RTK array screen, we identified the RTK AXL as a top upregulated RTK in response to smoke. Both expression and signaling activity of AXL were indeed elevated in lung fibroblasts exposed to tobacco smoke, whereas no significant change to the levels of a canonical AXL ligand, Gas6 (growth arrest-specific 6), was seen upon smoke treatment. Notably, we found that smoke-exposed human lung fibroblasts exhibited highly proliferative and invasive activities and were capable of inducing fibrotic lung lesions in mice. Conversely, genetic suppression of AXL in smoke-exposed fibroblasts cells led to suppression of AXL downstream pathways and aggressive phenotypes. We further demonstrated that AXL interacted with MARCKS (myristoylated alanine-rich C kinase substrate) and cooperated with MARCKS in regulating downstream signaling activity and fibroblast invasiveness. Pharmacological inhibition of AXL with AXL-specific inhibitor R428 showed selectivity for smoke-exposed fibroblasts. In all, our data suggest that AXL is a potential marker for smoke-associated PF and that targeting of the AXL pathway is a potential therapeutic strategy in treating tobacco smoking-related PF.
Collapse
Affiliation(s)
- David C. Yang
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| | - Shenwen Gu
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| | - Ji-Min Li
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| | - Ssu-Wei Hsu
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| | - Szu-Jung Chen
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| | - Wen-Hsin Chang
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Institute of Molecular Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Hsien Chen
- Division of Pulmonary and Critical Care Medicine, and Center for Comparative Respiratory Biology and Medicine, Department of Internal Medicine, and
- Division of Nephrology, Department of Internal Medicine, University of California, Davis, Davis, California; and
| |
Collapse
|
35
|
Ma J, Dong C, Wei S, Qiu M, Wu P, Ou C, Zhang B, Zhang X, Yan J, Zhang Q, Zhong N. Serum Cytokine Profiling Identifies Axl as a New Biomarker Candidate for Active Eosinophilic Granulomatosis With Polyangiitis. Front Mol Biosci 2021; 8:653461. [PMID: 33987203 PMCID: PMC8112820 DOI: 10.3389/fmolb.2021.653461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 04/06/2021] [Indexed: 11/14/2022] Open
Abstract
Background: Eosinophilic granulomatosis with polyangiitis (EGPA) prognosis is generally favorable and is treated with combined corticosteroids/immunosuppressor(s) therapy. However, disease flares increase the number of clinical visits. Therefore, discovering new serum biomarkers for early identification of active EGPA is crucial. Objective: To identify reliable serum biomarkers to measure EGPA activity. Methods: The expression of 160 proteins was compared in sera from 15 inactive and 13 active EGPA patients by antibody-based microarray. Network-based analysis identified patterns in the different groups. Differentially expressed proteins (DEPs) in active disease were identified, and the correlation between their serum levels and clinical parameters was assessed. DEPs were further analyzed for GO enrichment and KEGG pathways. Finally, DEP marker candidates were validated by ELISA and Bio-plex as well as against a second cohort of 22 inactive and 18 active EGPA patients. Results: The active group presented higher peripheral and sputum eosinophil counts, FeNO, and FEV1 (% predicted) (P < 0.05). Network-based analysis showed scattered expression patterns in active subjects, but no significant bias in inactive subjects. Significant differences were observed in serum levels of 19 candidate markers, all of which were higher in active EGPA (P < 0.05). KEGG analysis indicated that DEPs were mainly involved in the MAPK, PI3K-Akt, RAS and Rap1 related pathways. Nine out of 19 candidate markers were positively correlated with peripheral eosinophil counts including FGF-7, SCF, GDNF, β-NGF, IGFBP-4, Axl, PIGF, Insulin, NT-4, ErbB3, OPN and BMP-4 (r = 0.693, r = 0.692, r = 0.687, r = 0.683, r = 0.671, r = 0.606, r = 0.571, r = 0.570, r = 0.516, respectively; P < 0.05), while two, CD14 and MCP-3, were negatively correlated (r = −0.644 and r = −0.515; P < 0.05). The higher expression of Axl, OPN, HCC-4, GDNF, and MCP-3 in active EGPA subjects was confirmed by ELISA and Custom Multiplex Bio-plex analyses. Conclusion: The serum protein profiles were significantly different between active and inactive EGPA. The expression of the candidate proteins correlated with peripheral blood eosinophil count. Serum Axl, OPN, HCC-4, GDNF, and MCP-3 levels were consistently higher in active EGPA, independent of the assessment methods. Finally, Axl had the largest AUC, indicating that this cytokine may serve as novel biomarker for the diagnosis of active EGPA.
Collapse
Affiliation(s)
- Jianjuan Ma
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Pediatric Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Cong Dong
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shushan Wei
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Minzhi Qiu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Penghui Wu
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changxing Ou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bomeng Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xueyan Zhang
- School of Basic Medical Sciences, The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jie Yan
- The Second Affiliated Hospital, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- Department of Pathophysiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China.,Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
36
|
Alle Q, Le Borgne E, Milhavet O, Lemaitre JM. Reprogramming: Emerging Strategies to Rejuvenate Aging Cells and Tissues. Int J Mol Sci 2021; 22:3990. [PMID: 33924362 PMCID: PMC8070588 DOI: 10.3390/ijms22083990] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is associated with a progressive and functional decline of all tissues and a striking increase in many "age-related diseases". Although aging has long been considered an inevitable process, strategies to delay and potentially even reverse the aging process have recently been developed. Here, we review emerging rejuvenation strategies that are based on reprogramming toward pluripotency. Some of these approaches may eventually lead to medical applications to improve healthspan and longevity.
Collapse
Affiliation(s)
- Quentin Alle
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| | - Enora Le Borgne
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| | - Ollivier Milhavet
- IRMB, University of Montpellier, INSERM, CNRS, 34295 Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University of Montpellier, INSERM, 34295 Montpellier, France; (Q.A.); (E.L.B.)
| |
Collapse
|
37
|
DeBerge M, Glinton K, Subramanian M, Wilsbacher LD, Rothlin CV, Tabas I, Thorp EB. Macrophage AXL receptor tyrosine kinase inflames the heart after reperfused myocardial infarction. J Clin Invest 2021; 131:139576. [PMID: 33529176 DOI: 10.1172/jci139576] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Abstract
Tyro3, AXL, and MerTK (TAM) receptors are activated in macrophages in response to tissue injury and as such have been proposed as therapeutic targets to promote inflammation resolution during sterile wound healing, including myocardial infarction. Although the role of MerTK in cardioprotection is well characterized, the unique role of the other structurally similar TAMs, and particularly AXL, in clinically relevant models of myocardial ischemia/reperfusion infarction (IRI) is comparatively unknown. Utilizing complementary approaches, validated by flow cytometric analysis of human and murine macrophage subsets and conditional genetic loss and gain of function, we uncover a maladaptive role for myeloid AXL during IRI in the heart. Cross signaling between AXL and TLR4 in cardiac macrophages directed a switch to glycolytic metabolism and secretion of proinflammatory IL-1β, leading to increased intramyocardial inflammation, adverse ventricular remodeling, and impaired contractile function. AXL functioned independently of cardioprotective MerTK to reduce the efficacy of cardiac repair, but like MerTK, was proteolytically cleaved. Administration of a selective small molecule AXL inhibitor alone improved cardiac healing, which was further enhanced in combination with blockade of MerTK cleavage. These data support further exploration of macrophage TAM receptors as therapeutic targets for myocardial infarction.
Collapse
Affiliation(s)
- Matthew DeBerge
- Department of Pathology and.,Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Kristofor Glinton
- Department of Pathology and.,Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Manikandan Subramanian
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Lisa D Wilsbacher
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA.,Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Carla V Rothlin
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Ira Tabas
- Departments of Medicine, Pathology and Cell Biology, and Physiology, Columbia University Irving Medical Center, New York City, New York, USA
| | - Edward B Thorp
- Department of Pathology and.,Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
38
|
Rovati L, Kaneko N, Pedica F, Monno A, Maehara T, Perugino C, Lanzillotta M, Pecetta S, Stone JH, Doglioni C, Manfredi AA, Pillai S, Della-Torre E. Mer tyrosine kinase (MerTK) as a possible link between resolution of inflammation and tissue fibrosis in IgG4-related disease. Rheumatology (Oxford) 2021; 60:4929-4941. [PMID: 33512463 DOI: 10.1093/rheumatology/keab096] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/17/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVES IgG4-related disease (IgG4-RD) is a systemic fibro-inflammatory disorder characterized by a dysregulated resolution of inflammation and wound healing response that might develop after an apoptotic insult induced by cytotoxic T lymphocytes (CTLs). Mer receptor tyrosine kinase (MerTK) and its ligand Protein S (ProS1) have a pivotal role in the resolution of inflammation, being implicated in the clearance of apoptotic cells, quenching of the immune response and development of tissue fibrosis. In the present work we aimed to investigate a possible involvement of the MerTK signalling pathway in the pathogenesis of IgG4-RD and development of tissue fibrosis. METHODS MerTK and ProS1 expression patterns in IgG4-RD lesions were evaluated by immunohistochemistry and immunofluorescence studies. Circulating MerTK+ monocytes, soluble Mer and MerTK ligands were measured in the peripheral blood of IgG4-RD patients and healthy controls by flow cytometry and ELISA, respectively. RESULTS MerTK was highly expressed by macrophages infiltrating IgG4-RD lesions. MerTK+ macrophages were more abundant in IgG4-RD than in Sjögren syndrome and interacted with apoptotic cells and ProS1 expressing T and B lymphocytes. Moreover, they expressed the pro-fibrotic cytokine TGF-β and their numbers declined following rituximab induced disease remission. Circulating MerTK+ monocytes, soluble Mer and MerTK ligands were not increased in the peripheral blood of patients with IgG4-RD. CONCLUSIONS The MerTK-ProS1 axis is activated in IgG4-RD lesions, possibly leading to persistent stimulation of processes involved in the resolution of inflammation and tissue fibrosis.
Collapse
Affiliation(s)
- Lucrezia Rovati
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Naoki Kaneko
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Federica Pedica
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antonella Monno
- Autoimmunity and Vascular Inflammation Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Takashi Maehara
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Section of Oral and Maxillofacial Oncology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Cory Perugino
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marco Lanzillotta
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Simone Pecetta
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - John H Stone
- Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Claudio Doglioni
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelo A Manfredi
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Autoimmunity and Vascular Inflammation Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Shiv Pillai
- Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emanuel Della-Torre
- Università Vita-Salute San Raffaele, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Ragon Institute of MGH, MIT, and Harvard, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
39
|
Role of the Gas6/TAM System as a Disease Marker and Potential Drug Target. DISEASE MARKERS 2021; 2021:2854925. [PMID: 33532004 PMCID: PMC7834835 DOI: 10.1155/2021/2854925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022]
|
40
|
Barnes PJ, Anderson GP, Fagerås M, Belvisi MG. Chronic lung diseases: prospects for regeneration and repair. Eur Respir Rev 2021; 30:30/159/200213. [PMID: 33408088 PMCID: PMC9488945 DOI: 10.1183/16000617.0213-2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
COPD and idiopathic pulmonary fibrosis (IPF) together represent a considerable unmet medical need, and advances in their treatment lag well behind those of other chronic conditions. Both diseases involve maladaptive repair mechanisms leading to progressive and irreversible damage. However, our understanding of the complex underlying disease mechanisms is incomplete; with current diagnostic approaches, COPD and IPF are often discovered at an advanced stage and existing definitions of COPD and IPF can be misleading. To halt or reverse disease progression and achieve lung regeneration, there is a need for earlier identification and treatment of these diseases. A precision medicine approach to treatment is also important, involving the recognition of disease subtypes, or endotypes, according to underlying disease mechanisms, rather than the current “one-size-fits-all” approach. This review is based on discussions at a meeting involving 38 leading global experts in chronic lung disease mechanisms, and describes advances in the understanding of the pathology and molecular mechanisms of COPD and IPF to identify potential targets for reversing disease degeneration and promoting tissue repair and lung regeneration. We also discuss limitations of existing disease measures, technical advances in understanding disease pathology, and novel methods for targeted drug delivery. Treatment outcomes with COPD and IPF are suboptimal. Better understanding of the diseases, such as targetable repair mechanisms, may generate novel therapies, and earlier diagnosis and treatment is needed to stop or even reverse disease progression.https://bit.ly/2Ga8J1g
Collapse
Affiliation(s)
- Peter J Barnes
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Gary P Anderson
- Lung Health Research Centre, University of Melbourne, Melbourne, Australia
| | | | - Maria G Belvisi
- National Heart & Lung Institute, Imperial College London, London, UK.,Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
41
|
Khan T, Dasgupta S, Ghosh N, Chaudhury K. Proteomics in idiopathic pulmonary fibrosis: the quest for biomarkers. Mol Omics 2021; 17:43-58. [PMID: 33073811 DOI: 10.1039/d0mo00108b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating chronic progressive and fibrosing lung disease that culminates in the destruction of alveolar integrity and dismal prognosis. Its etiology is unknown and pathophysiology remains unclear. While great advances have been made in elucidating the pathogenesis mechanism, considerable gaps related to information on pathogenetic pathways and key protein targets involved in the clinical course of the disease exist. These issues need to be addressed for better clinical management of this highly challenging disease. Omics approach has revolutionized the entire area of disease understanding and holds promise in its translation to clinical biomarker discovery. This review outlines the contribution of proteomics towards identification of important biomarkers in IPF in terms of their clinical utility, i.e. prognosis, differential diagnosis, disease progression and treatment monitoring. The major dysregulated pathways associated with IPF are also discussed. Based on numerous proteomics studies on human and animal models, it is proposed that IPF pathogenesis involves complex interactions of several pathways such as oxidative stress, endoplasmic reticulum stress, unfolded protein response, coagulation system, inflammation, abnormal wounding, fibroblast proliferation, fibrogenesis and deposition of extracellular matrix. These pathways and their key path-changing mediators need further validation in large well-planned multi-centric trials at various geographical locations for successful development of clinical biomarkers of this confounding disease.
Collapse
Affiliation(s)
- Tila Khan
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Sanjukta Dasgupta
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Nilanjana Ghosh
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, 721302, India.
| |
Collapse
|
42
|
Landolt L, Spagnoli GC, Hertig A, Brocheriou I, Marti HP. Fibrosis and cancer: shared features and mechanisms suggest common targeted therapeutic approaches. Nephrol Dial Transplant 2020; 37:1024-1032. [PMID: 33280031 DOI: 10.1093/ndt/gfaa301] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
Epidemiological studies support a strong link between organ fibrosis and epithelial cancers. Moreover, clinical and experimental investigations consistently indicate that these diseases intertwine and share strikingly overlapping features. As a deregulated response to injury occurring in all body tissues, fibrosis is characterized by activation of fibroblasts and immune cells, contributing to progressive deposition of extracellular matrix (ECM) and inflammation. Cancers are driven by genetic alterations resulting in dysregulated cell survival, proliferation and dissemination. However, non-cancerous components of tumour tissues including fibroblasts, inflammatory cells and ECM play key roles in oncogenesis and cancer progression by providing a pro-mutagenic environment where cancer cells can develop, favouring their survival, expansion and invasiveness. Additional commonalities of fibrosis and cancer are also represented by overproduction of growth factors, like transforming growth factor β, epithelial-to-mesenchymal transition, high oxidative stress, Hippo pathway dysfunctions and enhanced cellular senescence. Here, we review advances in the analysis of cellular and molecular mechanisms involved in the pathogenesis of both organ fibrosis and cancer, with particular reference to chronic kidney diseases and renal cell cancers. Most importantly, improved understanding of common features is contributing to the development of innovative treatment strategies targeting shared mechanisms.
Collapse
Affiliation(s)
- Lea Landolt
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Giulio C Spagnoli
- National Research Council, Institute of Translational Pharmacology, Rome, Italy
| | - Alexandre Hertig
- Sorbonne Université, INSERM UMR S1155, Pitié-Salpêtrière Hospital, APHP6, Paris, France and
| | - Isabelle Brocheriou
- Sorbonne Université, INSERM UMR S1155, Pitié-Salpêtrière Hospital, APHP6, Paris, France and.,Department of Pathology, Pitié-Salpêtrière Hospital, Paris, France
| | - Hans-Peter Marti
- Department of Clinical Medicine, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
43
|
Li W, Xie L, Ma J, Cheng M, Fan L, Xu Y, Wang B, Chen W. Gas6 or Mer deficiency ameliorates silica-induced autophagosomes accumulation in mice lung. Toxicol Lett 2020; 337:28-37. [PMID: 33232774 DOI: 10.1016/j.toxlet.2020.11.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/14/2022]
Abstract
Published evidences have shown that autophagy plays an important role in silica-induced lung inflammation and collagen deposition. Our previous study found that the level of growth arrest-specific protein 6 (Gas6) in bronchoalveolar lavage fluid was increased after silica exposure. However, it is unclear whether Gas6 is involved in the regulation of silica-induced autophagy dysfunction. In this study, we observed an autophagosomes accumulation in wild-type C57BL/6 (WT) mice lung after silica intratracheal instillation and then investigated whether genetic loss of Gas6 (Gas6-/-) could ameliorate it. Our data showed that Gas6-/- mice exhibited a limited autophagosomes accumulation from days 7-84 after silica exposure, revealed by reduced induction and increased degradation of autophagosomes in mice lung tissue. Interestingly, silica particles could elevate the expression of Mer receptor, which was significantly decreased in Gas6-/- mice (P < 0.05). Furthermore, we found that Mer deficiency (Mer-/-) could also reduce the formation of autophagosomes and restore the function of impaired lysosomes in silica-treated mice. Taken together, our results indicate that genetic loss of Gas6 attenuates silica-induced autophagosomes accumulation partly through down-regulating the expression of Mer receptor. Targeting Gas6/Mer-mediated autophagy pathway may provide a novel insight into the prevention and therapy of silica-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Wei Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Li Xie
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jixuan Ma
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Man Cheng
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Lieyang Fan
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yiju Xu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Bin Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Weihong Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China; Key Laboratory of Environment and Health, Ministry of Education and Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
44
|
Di Stasi R, De Rosa L, D'Andrea LD. Therapeutic aspects of the Axl/Gas6 molecular system. Drug Discov Today 2020; 25:2130-2148. [PMID: 33002607 DOI: 10.1016/j.drudis.2020.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Axl receptor tyrosine kinase (RTK) and its ligand, growth arrest-specific protein 6 (Gas6), are involved in several biological functions and participate in the development and progression of a range of malignancies and autoimmune disorders. In this review, we present this molecular system from a drug discovery perspective, highlighting its therapeutic implications and challenges that need to be addressed. We provide an update on Axl/Gas6 axis biology, exploring its role in fields ranging from angiogenesis, cancer development and metastasis, immune response and inflammation to viral infection. Finally, we summarize the molecules that have been developed to date to target the Axl/Gas6 molecular system for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luca D D'Andrea
- Istituto di Biostrutture e Bioimmagini, CNR, Via Nizza 52, 10126 Torino, Italy.
| |
Collapse
|
45
|
Gas6/Axl Signaling Pathway in the Tumor Immune Microenvironment. Cancers (Basel) 2020; 12:cancers12071850. [PMID: 32660000 PMCID: PMC7408754 DOI: 10.3390/cancers12071850] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 01/19/2023] Open
Abstract
Receptor tyrosine kinases have been shown to dysregulate a number of pathways associated with tumor development, progression, and metastasis. Axl is a receptor tyrosine kinase expressed in many cancer types and has been associated with therapy resistance and poor clinical prognosis and outcomes. In addition, Axl and its ligand growth arrest specific 6 (Gas6) protein are expressed by a number of host cells. The Gas6/Axl signaling pathway has been implicated in the promotion of tumor cell proliferation, survival, migration, invasion, angiogenesis, and immune evasion. As a result, Axl is an attractive, novel therapeutic target to impair multiple stages of tumor progression from both neoplastic and host cell axes. This review focuses on the role of the Gas6/Axl signaling pathway in promoting the immunosuppressive tumor microenvironment, as immune evasion is considered one of the hallmarks of cancer. The review discusses the structure and activation of the Gas6/Axl signaling pathway, GAS6 and AXL expression patterns in the tumor microenvironment, mechanisms of Axl-mediated tumor immune response, and the role of Gas6/Axl signaling in immune cell recruitment.
Collapse
|
46
|
Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev 2020; 60:101063. [PMID: 32272170 DOI: 10.1016/j.arr.2020.101063] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/25/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Fibrosis is the final hallmark of pathological remodeling, which is a major contributor to the pathogenesis of various chronic diseases and aging-related organ failure to fully control chronic wound-healing and restoring tissue function. The process of fibrosis is involved in the pathogenesis of the kidney, lung, liver, heart and other tissue disorders. Wnt is a highly conserved signaling in the aberrant wound repair and fibrogenesis, and sustained Wnt activation is correlated with the pathogenesis of fibrosis. In particular, mounting evidence has revealed that Wnt signaling played important roles in cell fate determination, proliferation and cell polarity establishment. The expression and distribution of Wnt signaling in different tissues vary with age, and these changes have key effects on maintaining tissue homeostasis. In this review, we first describe the major constituents of the Wnt signaling and their regulation functions. Subsequently, we summarize the dysregulation of Wnt signaling in aging-related fibrotic tissues such as kidney, liver, lung and cardiac fibrosis, followed by a detailed discussion of its involvement in organ fibrosis. In addition, the crosstalk between Wnt signaling and other pathways has the potential to profoundly add to the complexity of organ fibrosis. Increasing studies have demonstrated that a number of Wnt inhibitors had the potential role against tissue fibrosis, specifically in kidney fibrosis and the implications of Wnt signaling in aging-related diseases. Therefore, targeting Wnt signaling might be a novel and promising therapeutic strategy against aging-related tissue fibrosis.
Collapse
Affiliation(s)
- He-He Hu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, California, 92897, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
47
|
Role of Gas6 and TAM Receptors in the Identification of Cardiopulmonary Involvement in Systemic Sclerosis and Scleroderma Spectrum Disorders. DISEASE MARKERS 2020; 2020:2696173. [PMID: 32454903 PMCID: PMC7240795 DOI: 10.1155/2020/2696173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 04/24/2020] [Indexed: 01/25/2023]
Abstract
Background Few biomarkers are available for early identification of pulmonary arterial hypertension (PAH) and interstitial lung disease (ILD) in systemic sclerosis (SS) and scleroderma spectrum disorders (SSD). Aims To evaluate Gas6, sAxl, and sMer as biomarkers for cardiopulmonary complications of SS and SSD. Methods In a cross-sectional observational study, we recruited 125 consecutive patients, affected by SS and SSD and referred to a tertiary-level pulmonary hypertension outpatient clinic. All patients underwent a comprehensive evaluation for identification of PAH and ILD. Gas6, sMer, and sAxl concentrations were measured with ELISA protocols, and concentrations were compared according to PAH or ILD. Results Nineteen subjects had pulmonary hypertension (PH) (14 PAH), and 39 had ILD (6 severe). Plasma sMer was increased in PAH (18.6 ng/ml IQR [11.7-20.3]) with respect to the absence (12.4 [8.0-15.8]) or other form of pulmonary hypertension (9.6 [7.4-12.5]; K–W variance p < 0.04). Conversely, Gas6 and sAxl levels were slightly increased in mild ILD (25.8 ng/ml [19.5-32.1] and 24.6 [20.1-32.5]) and reduced in severe ILD (16.6 [15.0-22.1] and 15.5 [14.9-22.4]) in comparison to no evidence of ILD (23.4 [18.8-28.1] and 21.6 [18.1-28.4]; K–W, p ≤ 0.05). Plasma sMer ≥ 19 ng/ml has 50% sensitivity and 92% specificity in PAH identification (area under the ROC curve (AUC) 0.697, p < 0.03). Values of Gas6 ≤ 24.5 ng/ml and of sAxl ≤ 15.5 ng/ml have 100% and 67% sensitivity and 47% and 86% specificity, respectively, in identifying severe ILD (Gas6 AUC 0.787, p < 0.001; sAxl AUC 0.705, p < 0.05). Conclusions The assay of Gas6 sAxl and sMer may be useful to help in the identification of PAH and ILD in SS and SSD patients. The Gas6/TAM system seems to be relevant in cardiopulmonary complications of SS and SSD and merits further investigations.
Collapse
|
48
|
Wang Y, Kuai Q, Gao F, Wang Y, He M, Zhou H, Han G, Jiang X, Ren S, Yu Q. Overexpression of TIM-3 in Macrophages Aggravates Pathogenesis of Pulmonary Fibrosis in Mice. Am J Respir Cell Mol Biol 2020; 61:727-736. [PMID: 31162951 DOI: 10.1165/rcmb.2019-0070oc] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive lung disorder and lacks effective treatments because of unclear mechanisms. Aberrant function of alveolar macrophages is directly linked to pulmonary fibrosis. Here, we show TIM-3 (T-cell immunoglobulin domain and mucin domain-3), a key regulator of macrophage function, aggravates pulmonary fibrosis. TIM-3 mRNA of patients with IPF was analyzed based on the Gene Expression Omnibus and Array Express databases. Lung pathology and profibrotic molecules were assessed in a bleomycin (BLM)-induced pulmonary fibrosis model using wild-type (WT) and TIM-3 transgenic (TIM-3-TG) mice. Macrophage cells, RAW264.7, were then applied to investigate the effect of macrophage TIM-3 under BLM exposure in vitro. Macrophage depletion and adoptive-transfer experiments were finally performed to examine lung morphology and profibrotic molecules. TIM-3 expression was increased both in patients with IPF and in our BLM-induced mouse model. TIM-3-TG mice developed more serious pathological changes in lung tissue and higher expressions of TGF-β1 (transforming growth factor-β1) and IL-10 than WT mice. After BLM treatment, TGF-β1 and IL-10 expression was significantly decreased in RAW264.7 cells after TIM-3 knock-out, whereas it was increased in TIM-3-TG peritoneal macrophages. The scores of pulmonary fibrosis in WT and TIM-3-TG mice were significantly reduced, and there was no difference between them after macrophage depletion. Furthermore, WT mice receiving adoptive macrophages from TIM-3-TG mice also had more serious lung fibrosis and increased expression of TGF-β1 and IL-10 than those receiving macrophages from WT mice. Our findings revealed that overexpressed TIM-3 in alveolar macrophages aggravated pulmonary fibrosis.
Collapse
Affiliation(s)
- Yu Wang
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Qiyuan Kuai
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Fenghua Gao
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Yanbing Wang
- Beijing Institute of Transfusion Medicine, Beijing, China.,School of Life Sciences, Jilin University, Changchun, China
| | - Min He
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Hong Zhou
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Gencheng Han
- Institute of Beijing Brain Sciences, Beijing, China; and
| | - Xingwei Jiang
- Beijing Institute of Transfusion Medicine, Beijing, China
| | - Suping Ren
- Beijing Institute of Transfusion Medicine, Beijing, China.,Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Qun Yu
- Beijing Institute of Transfusion Medicine, Beijing, China.,Beijing Advanced Innovation Centre for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| |
Collapse
|
49
|
Greiffo FR, Viteri-Alvarez V, Frankenberger M, Dietel D, Ortega-Gomez A, Lee JS, Hilgendorff A, Behr J, Soehnlein O, Eickelberg O, Fernandez IE. CX3CR1-fractalkine axis drives kinetic changes of monocytes in fibrotic interstitial lung diseases. Eur Respir J 2020; 55:13993003.00460-2019. [PMID: 31744836 DOI: 10.1183/13993003.00460-2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/31/2019] [Indexed: 12/21/2022]
Abstract
Circulating immune cell populations have been shown to contribute to interstitial lung disease (ILD). In this study, we analysed circulating and lung resident monocyte populations, and assessed their phenotype and recruitment from the blood to the lung in ILD. Flow cytometry analysis of blood samples for quantifying circulating monocytes was performed in 105 subjects: 83 with ILD (n=36, n=28 and n=19 for nonspecific interstitial pneumonia, hypersensitivity pneumonitis and connective-tissue disease-associated ILD, respectively), as well as 22 controls. Monocyte localisation and abundance were assessed using immunofluorescence and flow cytometry of lung tissue. Monocyte populations were cultured either alone or with endothelial cells to assess fractalkine-dependent transmigration pattern. We show that circulating classical monocytes (CM) were increased in ILD compared with controls, while nonclassical monocytes (NCM) were decreased. CM abundance correlated inversely with lung function, while NCM abundance correlated positively. Both CCL2 and CX3CL1 concentrations were increased in plasma and lungs of ILD patients. Fractalkine co-localised with ciliated bronchial epithelial cells, thereby creating a chemoattractant gradient towards the lung. Fractalkine enhanced endothelial transmigration of NCM in ILD samples only. Immunofluorescence, as well as flow cytometry, showed an increased presence of NCM in fibrotic niches in ILD lungs. Moreover, NCM in the ILD lungs expressed increased CX3CR1, M2-like and phagocytic markers. Taken together, our data support that in ILD, fractalkine drives the migration of CX3CR1+ NCM to the lungs, thereby perpetuating the local fibrotic process.
Collapse
Affiliation(s)
- Flavia R Greiffo
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Valeria Viteri-Alvarez
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marion Frankenberger
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Daniela Dietel
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Almudena Ortega-Gomez
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), Munich, Germany
| | - Joyce S Lee
- Division of Pulmonary Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Dept of Neonatalogy, Perinatal Center Grosshadern Ludwig-Maximilians University, Munich, Germany.,Center for Comprehensive Developmental Care, Dr von Haunersches Children's Hospital University, Hospital Ludwig-Maximilians University, Munich, Germany.,CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- Asklepios Fachkliniken München-Gauting, Munich, Germany.,Comprehensive Pneumology Center, Medizinische Klinik und Poliklinik V, Klinikum der Ludwig-Maximilians Universität (LMU), Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Oliver Soehnlein
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University (LMU), Munich, Germany.,Deutsches Zentrum für Herz-Kreislaufforschung (DZHK), partner site Munich Heart Alliance, Munich, Germany.,Dept of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Oliver Eickelberg
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Division of Pulmonary Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA.,These authors contributed equally
| | - Isis E Fernandez
- Comprehensive Pneumology Center, Ludwig-Maximilians University (LMU), University Hospital Grosshadern, and Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany .,CPC-M bioArchive, Helmholtz Zentrum München, Comprehensive Pneumology Center (CPC), Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,These authors contributed equally
| |
Collapse
|
50
|
Brenig R, Pop OT, Triantafyllou E, Geng A, Singanayagam A, Perez-Shibayama C, Besse L, Cupovic J, Künzler P, Boldanova T, Brand S, Semela D, Duong FHT, Weston CJ, Ludewig B, Heim MH, Wendon J, Antoniades CG, Bernsmeier C. Expression of AXL receptor tyrosine kinase relates to monocyte dysfunction and severity of cirrhosis. Life Sci Alliance 2019; 3:3/1/e201900465. [PMID: 31822557 PMCID: PMC6907389 DOI: 10.26508/lsa.201900465] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 11/07/2019] [Accepted: 12/02/2019] [Indexed: 12/14/2022] Open
Abstract
Infectious complications in patients with cirrhosis frequently initiate episodes of decompensation and substantially contribute to the high mortality. Mechanisms of the underlying immuneparesis remain underexplored. TAM receptors (TYRO3/AXL/MERTK) are important inhibitors of innate immune responses. To understand the pathophysiology of immuneparesis in cirrhosis, we detailed TAM receptor expression in relation to monocyte function and disease severity prior to the onset of acute decompensation. TNF-α/IL-6 responses to lipopolysaccharide were attenuated in monocytes from patients with cirrhosis (n = 96) compared with controls (n = 27) and decreased in parallel with disease severity. Concurrently, an AXL-expressing (AXL+) monocyte population expanded. AXL+ cells (CD14+CD16highHLA-DRhigh) were characterised by attenuated TNF-α/IL-6 responses and T cell activation but enhanced efferocytosis and preserved phagocytosis of Escherichia coli Their expansion correlated with disease severity, complications, infection, and 1-yr mortality. AXL+ monocytes were generated in response to microbial products and efferocytosis in vitro. AXL kinase inhibition and down-regulation reversed attenuated monocyte inflammatory responses in cirrhosis ex vivo. AXL may thus serve as prognostic marker and deserves evaluation as immunotherapeutic target in cirrhosis.
Collapse
Affiliation(s)
- Robert Brenig
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland,Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Oltin T Pop
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Evangelos Triantafyllou
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Anne Geng
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Arjuna Singanayagam
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Christian Perez-Shibayama
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Lenka Besse
- Laboratory of Experimental Oncology, Department of Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Jovana Cupovic
- Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Patrizia Künzler
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Tuyana Boldanova
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Stephan Brand
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - David Semela
- Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - François HT Duong
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Christopher J Weston
- Centre for Liver Research and National Institute for Health Research, Biomedical Research Unit, University of Birmingham, Birmingham, UK
| | - Burkhard Ludewig
- Institute of Immunobiology, Medical Research Centre, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Markus H Heim
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Julia Wendon
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK
| | - Charalambos G Antoniades
- Institute of Liver Studies, King’s College Hospital, King’s College London, London, UK,Hepatology Department, St. Mary’s Hospital, Imperial College London, London, UK
| | - Christine Bernsmeier
- Department of Biomedicine, University of Basel and University Centre for Gastrointestinal and Liver Diseases, Basel, Switzerland,Medical Research Centre and Division of Gastroenterology and Hepatology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland,Correspondence:
| |
Collapse
|