1
|
Shi H, Jiang M, Zhang X, Xia G, Shen X. Characteristics and food applications of aquatic collagen and its derivatives: A review. Food Res Int 2025; 202:115531. [PMID: 39967124 DOI: 10.1016/j.foodres.2024.115531] [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: 05/14/2024] [Revised: 12/05/2024] [Accepted: 12/28/2024] [Indexed: 02/20/2025]
Abstract
Collagen and its hydrolysates have high bioavailability, good biocompatibility, biodegradability, and biological activity which has meant that they have been widely used in food, medicine, cosmetics, and other industries. Although the properties and applications of collagen have been reviewed recently, few studies have focused on aquatic collagen. To provide readers with a deeper understanding of aquatic collagen, this review addresses the structure and properties of aquatic collagen and compares them with mammalian collagen, as well as the differences between collagen, gelatin, and collagen peptides. In contrast to mammalian collagen, aquatic collagen prevents zoonotic diseases, reduces environmental pollution, improves the utilization of aquatic resources, and facilitates the extraction and separation of active oligopeptides. Additionally, methods for screening functional peptides using in vitro digestion have been introduced. Finally, the review focuses on the applications of collagen and its derivatives in food preservation (packaging films, coatings, additives, and antifreeze peptides), drug delivery (microcapsules, emulsions, nanoparticles, and hydrogels), nutrition, and healthcare.
Collapse
Affiliation(s)
- Haohao Shi
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Key Laboratory of Seafood Processing of Haikou, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources of MOE, School of Food Science and Technology, Hainan University, Hainan 570228, China
| | - Mengqi Jiang
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Key Laboratory of Seafood Processing of Haikou, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources of MOE, School of Food Science and Technology, Hainan University, Hainan 570228, China
| | - Xueying Zhang
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Key Laboratory of Seafood Processing of Haikou, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources of MOE, School of Food Science and Technology, Hainan University, Hainan 570228, China
| | - Guanghua Xia
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Key Laboratory of Seafood Processing of Haikou, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources of MOE, School of Food Science and Technology, Hainan University, Hainan 570228, China.
| | - Xuanri Shen
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Key Laboratory of Seafood Processing of Haikou, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Engineering Research Center of Utilization of Tropical Polysaccharide Resources of MOE, School of Food Science and Technology, Hainan University, Hainan 570228, China; College of Food Science and Technology, Hainan Tropical Ocean University, Sanya 572022, China.
| |
Collapse
|
2
|
Khan MM, Galea G, Jung J, Zukowska J, Lauer D, Tuechler N, Halavatyi A, Tischer C, Haberkant P, Stein F, Jung F, Landry JJM, Khan AM, Oorschot V, Becher I, Neumann B, Muley T, Winter H, Duerr J, Mall MA, Grassi A, de la Cueva E, Benes V, Gote-Schniering J, Savitski M, Pepperkok R. Dextromethorphan inhibits collagen and collagen-like cargo secretion to ameliorate lung fibrosis. Sci Transl Med 2024; 16:eadj3087. [PMID: 39693409 DOI: 10.1126/scitranslmed.adj3087] [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: 06/19/2023] [Revised: 04/25/2024] [Accepted: 11/01/2024] [Indexed: 12/20/2024]
Abstract
Excessive deposition of fibrillar collagen in the interstitial extracellular matrix (ECM) of human lung tissue causes fibrosis, which can ultimately lead to organ failure. Despite our understanding of the molecular mechanisms underlying the disease, no cure for pulmonary fibrosis has yet been found. We screened a drug library and found that dextromethorphan (DXM), a cough expectorant, reduced the amount of excess fibrillar collagen deposited in the ECM in cultured primary human lung fibroblasts, a bleomycin mouse model, and a cultured human precision-cut lung slice model of lung fibrosis. The reduced extracellular fibrillar collagen upon DXM treatment was due to reversible trafficking inhibition of collagen type I (COL1) in the endoplasmic reticulum (ER) in TANGO1- and HSP47-positive structures. Mass spectrometric analysis showed that DXM promoted hyperhydroxylation of proline and lysine residues on various collagens (COL1, COL3, COL4, COL5, COL7, and COL12) and latent transforming growth factor-β-binding protein (LTBP1 and LTBP2) peptides, coinciding with their secretion block. Additionally, proteome profiling of DXM-treated cells showed increased thermal stability of prolyl-hydroxylases P3H2, P3H3, P3H4, P4HA1, and P4HA2, suggesting a change in their activity. Transcriptome analysis of profibrotic stimulated primary human lung fibroblasts and human ex vivo lung slices after DXM treatment showed activation of an antifibrotic program through regulation of multiple pathways, including the MMP-ADAMTS axis, WNT signaling, and fibroblast-to-myofibroblast differentiation. Together, these data obtained from in vitro, in vivo, and ex vivo models of lung fibrogenesis show that DXM has the potential to limit fibrosis through inhibition of COL1 membrane trafficking in the ER.
Collapse
Affiliation(s)
- Muzamil M Khan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - George Galea
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Juan Jung
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Joanna Zukowska
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - David Lauer
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Nadine Tuechler
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory and Heidelberg University, 69117 Heidelberg, Germany
- Institute for Computational Biomedicine (ICB), Faculty of Medicine, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Aliaksandr Halavatyi
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Christian Tischer
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Per Haberkant
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Ferris Jung
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Arif M Khan
- Centre for Bioimage Analysis, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Viola Oorschot
- Electron Microscopy Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Isabelle Becher
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Beate Neumann
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Translational Research Unit/Lung Biobank, Thoraxklinik, University Hospital Heidelberg, 69117 Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Translational Research Unit/Lung Biobank, Thoraxklinik, University Hospital Heidelberg, 69117 Heidelberg, Germany
| | - Julia Duerr
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin, 13353 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin, 13353 Berlin, Germany
- German Center for Lung Research (DZL), Associated Partner Site, 13353 Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 10178 Berlin, Germany
| | - Alessandro Grassi
- Laboratory Animal Resources, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Ernesto de la Cueva
- Laboratory Animal Resources, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Janine Gote-Schniering
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Genome Biology Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Advanced Light Microscopy Facility, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| |
Collapse
|
3
|
Huang HY, Zhang FW, Yu J, Xiao YH, Zhu D, Yi X, Lin X, Jin M, Jin HY, Huang YS, Ren SW. A computational analysis of the oncogenic and anti-tumor immunity role of P4HA3 in human cancers. PLoS Comput Biol 2024; 20:e1012284. [PMID: 39504328 PMCID: PMC11573185 DOI: 10.1371/journal.pcbi.1012284] [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] [Received: 07/04/2024] [Revised: 11/18/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Prolyl-4-hydroxylase subunit alpha3 (P4HA3) is a triple helical procollagen synthesis protein. The role of P4HA3 in cancer development is not well known and lacks comprehensive analyses among human cancers. This study aimed to investigate the relationship between P4HA3 expression and anti-tumor immunity and its prognostic value in pan-cancer. P4HA3 expression was analyzed from TIMER2.0, GTEx, GEPIA2.0 and TCGA databases. Genetic and DNA methylation alterations, survival analysis and proteins co-expression analysis of P4HA3 in cBio Cancer Genomics Portal, TCGA, GSCA and TIMER2.0. The correlation between P4HA3 expression and immune infiltration was analyzed by TIDE, XCELL, MCPCOUNTER, and EPIC. We performed EdU and transwell experiments to evaluate the influence of P4HA3 on the proliferation, migration and invasion abilities of different tumors. Patients derived xenograft (PDX) and subcutaneous transplantation models were utilized to explore the correlation between P4HA3 and immunotherapy response in triple-negative breast cancer (TNBC). Among 33 types of cancers, P4HA3 had generally different expression between different tumors, further analysis showed that the expression of P4HA3 was correlated with the cells infiltration of the tumor microenvironment (TME). The expression of P4HA3 was positively with the cell proliferation markers and epithelial-mesenchymal transition (EMT) markers. Moreover, P4HA3 deficiency inhibited the proliferation, migration and invasion abilities of tumor cells, and promoted anti-tumor immunotherapy of PD-1/PD-L1 inhibitor. This pan-cancer analysis of P4HA3 provides a comprehensive understanding of its oncogenic and prognosis role in different cancers, P4HA3 abnormal expression could be a useful biomarker for predicting the effectiveness of immunotherapy in cancer patients.
Collapse
Affiliation(s)
- Hong Yan Huang
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Fu Wei Zhang
- Department of Thoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Jie Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Yan Hong Xiao
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Di Zhu
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - XiaoLin Yi
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - XiaoHua Lin
- Department of Breast Surgery, Zhujiang Hospital, Southern Medical University, Guangdong, China
| | - Ming Jin
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Guangdong, China
| | - Hai Yun Jin
- Department of Gynaecology and Obstetrics, Southern Hospital Taihe Branch, Southern Medical University, Guangdong, China
| | - Yong Sheng Huang
- Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong, China
| | - Shu Wei Ren
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| |
Collapse
|
4
|
Bell JA, Davies ER, Brereton CJ, Vukmirovic M, Roberts JJW, Lunn K, Wickens L, Conforti F, Ridley RA, Ceccato J, Sayer LN, Johnston DA, Vallejo AF, Alzetani A, Jogai S, Marshall BG, Fabre A, Richeldi L, Monk PD, Skipp P, Kaminski N, Offer E, Wang Y, Davies DE, Jones MG. Spatial transcriptomic validation of a biomimetic model of fibrosis enables re-evaluation of a therapeutic antibody targeting LOXL2. Cell Rep Med 2024; 5:101695. [PMID: 39173635 PMCID: PMC11524965 DOI: 10.1016/j.xcrm.2024.101695] [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: 07/10/2023] [Revised: 02/26/2024] [Accepted: 07/29/2024] [Indexed: 08/24/2024]
Abstract
Matrix stiffening by lysyl oxidase-like 2 (LOXL2)-mediated collagen cross-linking is proposed as a core feedforward mechanism that promotes fibrogenesis. Failure in clinical trials of simtuzumab (the humanized version of AB0023, a monoclonal antibody against human LOXL2) suggested that targeting LOXL2 may not have disease relevance; however, target engagement was not directly evaluated. We compare the spatial transcriptome of active human lung fibrogenesis sites with different human cell culture models to identify a disease-relevant model. Within the selected model, we then evaluate AB0023, identifying that it does not inhibit collagen cross-linking or reduce tissue stiffness, nor does it inhibit LOXL2 catalytic activity. In contrast, it does potently inhibit angiogenesis consistent with an alternative, non-enzymatic mechanism of action. Thus, AB0023 is anti-angiogenic but does not inhibit LOXL2 catalytic activity, collagen cross-linking, or tissue stiffening. These findings have implications for the interpretation of the lack of efficacy of simtuzumab in clinical trials of fibrotic diseases.
Collapse
Affiliation(s)
- Joseph A Bell
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK
| | - Elizabeth R Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - Christopher J Brereton
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK
| | - Milica Vukmirovic
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | | | | | - Leanne Wickens
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - Franco Conforti
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK
| | - Robert A Ridley
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK
| | - Jessica Ceccato
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; Department of Medicine, University of Padova, Padova, Italy
| | - Lucy N Sayer
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK
| | - David A Johnston
- Biomedical Imaging Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andres F Vallejo
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK
| | - Aiman Alzetani
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; University Hospital Southampton, SO16 6YD Southampton, UK
| | - Sanjay Jogai
- University Hospital Southampton, SO16 6YD Southampton, UK
| | - Ben G Marshall
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; University Hospital Southampton, SO16 6YD Southampton, UK
| | - Aurelie Fabre
- Department of Histopathology, St. Vincent's University Hospital & UCD School of Medicine, University College Dublin, Dublin, Ireland
| | - Luca Richeldi
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Unità Operativa Complessa di Pneumologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli, Rome, Italy
| | | | - Paul Skipp
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, SO17 1BJ Southampton, UK; University Hospital Southampton, SO16 6YD Southampton, UK
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Emily Offer
- Medicines Discovery Catapult, Alderley Edge, UK
| | - Yihua Wang
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, SO17 1BJ Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - Donna E Davies
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - Mark G Jones
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; NIHR Southampton Biomedical Research Centre, University Hospital Southampton, SO16 6YD Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ Southampton, UK.
| |
Collapse
|
5
|
Song D, Tang X, Du J, Tao K, Li Y. Diazepam inhibits LPS-induced pyroptosis and inflammation and alleviates pulmonary fibrosis in mice by regulating the let-7a-5p/MYD88 axis. PLoS One 2024; 19:e0305409. [PMID: 38875245 PMCID: PMC11178199 DOI: 10.1371/journal.pone.0305409] [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] [Received: 02/28/2024] [Accepted: 05/29/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Pulmonary fibrosis caused by lung injury is accompanied by varying degrees of inflammation, and diazepam can reduce the levels of inflammatory factors. Therefore, the purpose of this study was to determine whether diazepam can inhibit inflammation and ameliorate pulmonary fibrosis by regulating the let-7a-5p/myeloid differentiation factor 88 (MYD88) axis. METHODS Lipopolysaccharide (LPS) was used to induce cell pyroptosis in an animal model of pulmonary fibrosis. After treatment with diazepam, changes in cell proliferation and apoptosis were observed, and the occurrence of inflammation and pulmonary fibrosis in the mice was detected. RESULTS The results showed that LPS can successfully induce cell pyroptosis and inflammatory responses and cause lung fibrosis in mice. Diazepam inhibits the expression of pyroptosis-related factors and inflammatory factors; moreover, it attenuates the occurrence of pulmonary fibrosis in mice. Mechanistically, diazepam can upregulate the expression of let-7a-5p, inhibit the expression of MYD88, and reduce inflammation and inhibit pulmonary fibrosis by regulating the let-7a-5p/MYD88 axis. CONCLUSION Our findings indicated that diazepam can inhibit LPS-induced pyroptosis and inflammatory responses and alleviate pulmonary fibrosis in mice by regulating the let-7a-5p/MYD88 axis.
Collapse
Affiliation(s)
- Duanyi Song
- Department of Anesthesiology, The Second People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Xuefang Tang
- Department of Anesthesiology, The Second People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Juan Du
- Department of Anesthesiology, The Second People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Kang Tao
- Department of Anesthesiology, The Second People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Yun Li
- Department of Anesthesiology, The Second People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| |
Collapse
|
6
|
Revert-Ros F, Ventura I, Prieto-Ruiz JA, Hernández-Andreu JM, Revert F. The Versatility of Collagen in Pharmacology: Targeting Collagen, Targeting with Collagen. Int J Mol Sci 2024; 25:6523. [PMID: 38928229 PMCID: PMC11203716 DOI: 10.3390/ijms25126523] [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: 05/08/2024] [Revised: 06/01/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
Collagen, a versatile family of proteins with 28 members and 44 genes, is pivotal in maintaining tissue integrity and function. It plays a crucial role in physiological processes like wound healing, hemostasis, and pathological conditions such as fibrosis and cancer. Collagen is a target in these processes. Direct methods for collagen modulation include enzymatic breakdown and molecular binding approaches. For instance, Clostridium histolyticum collagenase is effective in treating localized fibrosis. Polypeptides like collagen-binding domains offer promising avenues for tumor-specific immunotherapy and drug delivery. Indirect targeting of collagen involves regulating cellular processes essential for its synthesis and maturation, such as translation regulation and microRNA activity. Enzymes involved in collagen modification, such as prolyl-hydroxylases or lysyl-oxidases, are also indirect therapeutic targets. From another perspective, collagen is also a natural source of drugs. Enzymatic degradation of collagen generates bioactive fragments known as matrikines and matricryptins, which exhibit diverse pharmacological activities. Overall, collagen-derived peptides present significant therapeutic potential beyond tissue repair, offering various strategies for treating fibrosis, cancer, and genetic disorders. Continued research into specific collagen targeting and the application of collagen and its derivatives may lead to the development of novel treatments for a range of pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Fernando Revert
- Mitochondrial and Molecular Medicine Research Group, Facultad de Medicina y Ciencias de la Salud, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain; (F.R.-R.); (I.V.); (J.A.P.-R.); (J.M.H.-A.)
| |
Collapse
|
7
|
Jang J, Bentsen M, Kim YJ, Kim E, Garg V, Cai CL, Looso M, Li D. Endocardial HDAC3 is required for myocardial trabeculation. Nat Commun 2024; 15:4166. [PMID: 38755146 PMCID: PMC11099086 DOI: 10.1038/s41467-024-48362-6] [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: 04/16/2023] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Failure of proper ventricular trabeculation is often associated with congenital heart disease. Support from endocardial cells, including the secretion of extracellular matrix and growth factors is critical for trabeculation. However, it is poorly understood how the secretion of extracellular matrix and growth factors is initiated and regulated by endocardial cells. We find that genetic knockout of histone deacetylase 3 in the endocardium in mice results in early embryo lethality and ventricular hypotrabeculation. Single cell RNA sequencing identifies significant downregulation of extracellular matrix components in histone deacetylase 3 knockout endocardial cells. Secretome from cultured histone deacetylase 3 knockout mouse cardiac endothelial cells lacks transforming growth factor ß3 and shows significantly reduced capacity in stimulating cultured cardiomyocyte proliferation, which is remarkably rescued by transforming growth factor ß3 supplementation. Mechanistically, we identify that histone deacetylase 3 knockout induces transforming growth factor ß3 expression through repressing microRNA-129-5p. Our findings provide insights into the pathogenesis of congenital heart disease and conceptual strategies to promote myocardial regeneration.
Collapse
Affiliation(s)
- Jihyun Jang
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43215, USA
| | - Mette Bentsen
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Ye Jun Kim
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Erick Kim
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Vidu Garg
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43215, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43215, USA
| | - Chen-Leng Cai
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46201, USA
| | - Mario Looso
- Bioinformatics Core Unit (BCU), Max Planck Institute for Heart and Lung Research, 61231, Bad Nauheim, Germany
| | - Deqiang Li
- Center for Cardiovascular Research, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH, 43215, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, 43215, USA.
| |
Collapse
|
8
|
Russell JJ, Mummidi S, DeMarco VG, Grisanti LA, Bailey CA, Bender SB, Chandrasekar B. Integrated miRNA-mRNA networks underlie attenuation of chronic β-adrenergic stimulation-induced cardiac remodeling by minocycline. Physiol Genomics 2024; 56:360-366. [PMID: 38314697 PMCID: PMC11283891 DOI: 10.1152/physiolgenomics.00140.2023] [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: 11/14/2023] [Revised: 01/18/2024] [Accepted: 02/01/2024] [Indexed: 02/07/2024] Open
Abstract
Adverse cardiac remodeling contributes to heart failure development and progression, partly due to inappropriate sympathetic nervous system activation. Although β-adrenergic receptor (β-AR) blockade is a common heart failure therapy, not all patients respond, prompting exploration of alternative treatments. Minocycline, an FDA-approved antibiotic, has pleiotropic properties beyond antimicrobial action. Recent evidence suggests it may alter gene expression via changes in miRNA expression. Thus, we hypothesized that minocycline could prevent adverse cardiac remodeling induced by the β-AR agonist isoproterenol, involving miRNA-mRNA transcriptome alterations. Male C57BL/6J mice received isoproterenol (30 mg/kg/day sc) or vehicle via osmotic minipump for 21 days, along with daily minocycline (50 mg/kg ip) or sterile saline. Isoproterenol induced cardiac hypertrophy without altering cardiac function, which minocycline prevented. Total mRNA sequencing revealed isoproterenol altering gene networks associated with inflammation and metabolism, with fibrosis activation predicted by integrated miRNA-mRNA sequencing, involving miR-21, miR-30a, miR-34a, miR-92a, and miR-150, among others. Conversely, the cardiac miRNA-mRNA transcriptome predicted fibrosis inhibition in minocycline-treated mice, involving antifibrotic shifts in Atf3 and Itgb6 gene expression associated with miR-194 upregulation. Picrosirius red staining confirmed isoproterenol-induced cardiac fibrosis, prevented by minocycline. These results demonstrate minocycline's therapeutic potential in attenuating adverse cardiac remodeling through miRNA-mRNA-dependent mechanisms, especially in reducing cardiac fibrosis. NEW & NOTEWORTHY We demonstrate that minocycline treatment prevents cardiac hypertrophy and fibrotic remodeling induced by chronic β-adrenergic stimulation by inducing antifibrotic shifts in the cardiac miRNA-mRNA transcriptome.
Collapse
Affiliation(s)
- Jacob J Russell
- Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Srinivas Mummidi
- Health and Behavior Sciences, Texas A&M University-San Antonio, San Antonio, Texas, United States
| | - Vincent G DeMarco
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
- Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Laurel A Grisanti
- Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
| | - Chastidy A Bailey
- Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
| | - Shawn B Bender
- Biomedical Sciences, University of Missouri, Columbia, Missouri, United States
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
- Dalton Cardiovascular Center, University of Missouri, Columbia, Missouri, United States
| | - Bysani Chandrasekar
- Research Service, Harry S. Truman Memorial Veterans Hospital, Columbia, Missouri, United States
- Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
- Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, United States
- Dalton Cardiovascular Center, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
9
|
Jang J, Bentsen M, Kim YJ, Kim E, Garg V, Cai CL, Looso M, Li D. Endocardial HDAC3 is required for myocardial trabeculation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.12.536668. [PMID: 37886504 PMCID: PMC10602027 DOI: 10.1101/2023.04.12.536668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
BACKGROUND Trabeculation, a key process in early heart development, is the formation of myocardial trabecular meshwork. The failure of trabeculation often leads to embryonic lethality. Support from endocardial cells, including the secretion of extracellular matrix (ECM) and growth factors is critical for trabeculation; however, it is unknown how the secretion of ECM and growth factors is initiated and regulated by endocardial cells. METHODS Various cellular and mouse models in conjunction with biochemical and molecular tools were employed to study the role of histone deacetylase 3 (HDAC3) in the developing endocardium. RESULTS We found that genetic deletion of Hdac3 in endocardial cells in mice resulted in early embryo lethality presenting as a hypotrabeculation cardiac phenotype. Single cell RNA sequencing identified several ECM components including collagens that were significantly downregulated in Hdac3 knockout (KO) endocardial cells. When cultured with supernatant from Hdac3 KO mouse cardiac endothelial cells (MCECs), wild-type mouse embryonic cardiomyocytes showed decreased proliferation, suggesting that growth signaling from Hdac3 KO MCECs is disrupted. Subsequent transcriptomic analysis revealed that transforming growth factor β3 (TGFβ3) was significantly downregulated in Hdac3 KO MCECs and Hdac3 cardiac endothelial KO hearts. Mechanistically, we identified that microRNA (miR)-129-5p was significantly upregulated in Hdac3 KO MCECs and Hdac3 cardiac endothelial KO hearts. Overexpression of miR-129-5p repressed Tgfβ3 expression in wild-type MCECs, whereas knockdown of miR-129-5p restored Tgfβ3 expression in Hdac3 KO MCECs. CONCLUSION Our findings reveal a critical signaling pathway in which endocardial HDAC3 promotes trabecular myocardium growth by stimulating TGFβ signaling through repressing miR-129-5p, providing novel insights into the etiology of congenital heart disease and conceptual strategies to promote myocardial regeneration.
Collapse
|
10
|
Huang X, Sun W, Nie B, Li JJ, Jing F, Zhou XL, Ni XY, Ni XC. Adipose-derived stem cells repair radiation-induced chronic lung injury via inhibiting TGF-β1/Smad 3 signaling pathway. Open Med (Wars) 2023; 18:20230850. [PMID: 38025537 PMCID: PMC10655693 DOI: 10.1515/med-2023-0850] [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: 06/29/2023] [Revised: 10/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
To investigate the effect of adipose-derived stem cells (ASCs) transplantation on radiation-induced lung injury (RILI), Sprague-Dawley rats were divided into phosphate-buffered saline (PBS) group, ASCs group, Radiation + PBS group, and Radiation + ASCs group. Radiation + PBS and Radiation + ASCs groups received single dose of 30 Gy X-ray radiation to the right chest. The Radiation + PBS group received 1 mL PBS suspension and Radiation + ASCs group received 1 mL PBS suspension containing 1 × 107 CM-Dil-labeled ASCs. The right lung tissue was collected on Days 30, 90, and 180 after radiation. Hematoxylin-eosin and Masson staining were performed to observe the pathological changes and collagen fiber content in the lung tissue. Immunohistochemistry (IHC) and western blot (WB) were used to detect levels of fibrotic markers collagen I (Collal), fibronectin (FN), as well as transforming growth factor-β1 (TGF-β1), p-Smad 3, and Smad 3. Compared with the non-radiation groups, the radiation groups showed lymphocyte infiltration on Day 30 after irradiation and thickened incomplete alveolar walls, collagen deposition, and fibroplasia on Days 90 and 180. ASCs relieved these changes on Day 180 (Masson staining, P = 0.0022). Compared with Radiation + PBS group, on Day 180 after irradiation, the Radiation + ASCs group showed that ASCs could significantly decrease the expressions of fibrosis markers Collal (IHC: P = 0.0022; WB: P = 0.0087) and FN (IHC: P = 0.0152; WB: P = 0.026) and inhibit the expressions of TGF-β1 (IHC: P = 0.026; WB: P = 0.0152) and p-Smad 3 (IHC: P = 0.0043; WB: P = 0.0087) in radiation-induced injured lung tissue. These indicated that ASCs could relieve RILI by inhibiting TGF-β1/Smad 3 signaling pathway.
Collapse
Affiliation(s)
- Xin Huang
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Wei Sun
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Bin Nie
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Juan-juan Li
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Fei Jing
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Xiao-li Zhou
- Department of Pathology, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Xin-ye Ni
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, Jiangsu, 213000, China
| | - Xin-chu Ni
- Department of Radiotherapy, The Affiliated Changzhou Second People’s Hospital of Nanjing Medical University, Changzhou Second People’s Hospital, Changzhou Medical Center, Nanjing Medical University, No. 68, Gehuzhonglu Road, Wujin District, Changzhou, Jiangsu, 213000, China
| |
Collapse
|
11
|
Zhang J, Jiang X, Li X, Sun H, Wang M, Zhang W, Li H, Wang H, Zhuang M, Zhang L, Lu L, Tang J. Pulmonary Toxicity Assessment after a Single Intratracheal Inhalation of Chlorhexidine Aerosol in Mice. TOXICS 2023; 11:910. [PMID: 37999562 PMCID: PMC10675078 DOI: 10.3390/toxics11110910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023]
Abstract
Guanidine disinfectants are important chemical agents with a broad spectrum of activity that are effective against most microorganisms. Chlorhexidine, one of the most used guanidine disinfectants, is added to shampoo and mouthwash and applied in medical device sterilization. During the use of chlorhexidine, aerosols with micron particle size may be formed, which may cause inhalation toxicity. To assess the toxicity of inhaled chlorhexidine aerosol, mice underwent the intratracheal instillation of different concentrations of chlorhexidine (0, 0.125%, 0.25%, 0.5%, and 1%) using a MicroSprayer Aerosolizer. The mice were exposed for eight weeks and then sacrificed to obtain lung tissue for subsequent experiments. Histopathology staining revealed damaged lung tissues and increased collagen exudation. At the same time, pulmonary function tests showed that chlorhexidine exposure could cause restrictive ventilatory dysfunction, consistent with pulmonary fibrosis. The results of transcriptome analyses suggest that chlorhexidine may trigger an inflammatory response and promote the activation of pathways related to extracellular matrix deposition. Further, we identified that chlorhexidine exposure might enhance mucus secretion by up-regulating Muc5b and Muc5ac genes, thereby inducing fibrosis-like injury. These findings underscore the need for standardized use of disinfectants and the assessment of their inhalation toxicity.
Collapse
Affiliation(s)
- Jianzhong Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; (J.Z.); (X.J.); (L.Z.)
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Xinmin Jiang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; (J.Z.); (X.J.); (L.Z.)
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Xin Li
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - He Sun
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Mingyue Wang
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Wanjun Zhang
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Haonan Li
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Hongmei Wang
- Department of Respiratory Medicine, Affiliated Hospital of Medical College of Qingdao University, Qingdao 266021, China; (H.W.); (M.Z.)
| | - Min Zhuang
- Department of Respiratory Medicine, Affiliated Hospital of Medical College of Qingdao University, Qingdao 266021, China; (H.W.); (M.Z.)
| | - Lin Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; (J.Z.); (X.J.); (L.Z.)
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Lin Lu
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; (J.Z.); (X.J.); (L.Z.)
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| | - Jinglong Tang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250001, China; (J.Z.); (X.J.); (L.Z.)
- Department of Environmental and Occupational Health, School of Public Health, Qingdao University, Qingdao 266071, China; (X.L.); (H.S.); (M.W.); (W.Z.); (H.L.)
| |
Collapse
|
12
|
Hayton C, Ahmed W, Cunningham P, Piper-Hanley K, Pearmain L, Chaudhuri N, Leonard C, Blaikley JF, Fowler SJ. Changes in lung epithelial cell volatile metabolite profile induced by pro-fibrotic stimulation with TGF- β1. J Breath Res 2023; 17:046012. [PMID: 37619557 DOI: 10.1088/1752-7163/acf391] [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: 04/17/2023] [Accepted: 08/24/2023] [Indexed: 08/26/2023]
Abstract
Volatile organic compounds (VOCs) have shown promise as potential biomarkers in idiopathic pulmonary fibrosis. Measuring VOCs in the headspace ofin vitromodels of lung fibrosis may offer a method of determining the origin of those detected in exhaled breath. The aim of this study was to determine the VOCs associated with two lung cell lines (A549 and MRC-5 cells) and changes associated with stimulation of cells with the pro-fibrotic cytokine, transforming growth factor (TGF)-β1. A dynamic headspace sampling method was used to sample the headspace of A549 cells and MRC-5 cells. These were compared to media control samples and to each other to identify VOCs which discriminated between cell lines. Cells were then stimulated with the TGF-β1 and samples were compared between stimulated and unstimulated cells. Samples were analysed using thermal desorption-gas chromatography-mass spectrometry and supervised analysis was performed using sparse partial least squares-discriminant analysis (sPLS-DA). Supervised analysis revealed differential VOC profiles unique to each of the cell lines and from the media control samples. Significant changes in VOC profiles were induced by stimulation of cell lines with TGF-β1. In particular, several terpenoids (isopinocarveol, sativene and 3-carene) were increased in stimulated cells compared to unstimulated cells. VOC profiles differ between lung cell lines and alter in response to pro-fibrotic stimulation. Increased abundance of terpenoids in the headspace of stimulated cells may reflect TGF-β1 cell signalling activity and metabolic reprogramming. This may offer a potential biomarker target in exhaled breath in IPF.
Collapse
Affiliation(s)
- Conal Hayton
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Waqar Ahmed
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Peter Cunningham
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Karen Piper-Hanley
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Laurence Pearmain
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Nazia Chaudhuri
- School of Medicine, The University of Ulster, Magee Campus, Londonderry, United Kingdom
| | - Colm Leonard
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - John F Blaikley
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Stephen J Fowler
- Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
- NIHR-Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| |
Collapse
|
13
|
Waldrep KM, Rodgers JI, Garrett SM, Wolf BJ, Feghali-Bostwick CA. The Role of SOX9 in IGF-II-Mediated Pulmonary Fibrosis. Int J Mol Sci 2023; 24:11234. [PMID: 37510994 PMCID: PMC10378869 DOI: 10.3390/ijms241411234] [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: 03/22/2023] [Revised: 06/29/2023] [Accepted: 07/02/2023] [Indexed: 07/30/2023] Open
Abstract
Pulmonary fibrosis (PF) associated with systemic sclerosis (SSc) results in significant morbidity and mortality. We previously reported that insulin-like growth factor-II (IGF-II) is overexpressed in lung tissues and fibroblasts from SSc patients, and IGF-II fosters fibrosis by upregulating collagen type I, fibronectin, and TGFβ. We now show that IGF-II augments mRNA levels of profibrotic signaling molecules TGFβ2 (p ≤ 0.01) and TGFβ3 (p ≤ 0.05), collagen type III (p ≤ 0.01), and the collagen posttranslational modification enzymes P4HA2 (p ≤ 0.05), P3H2 (p ≤ 0.05), LOX (p = 0.065), LOXL2 (p ≤ 0.05), LOXL4 (p ≤ 0.05) in primary human lung fibroblasts. IGF-II increases protein levels of TGFβ2 (p ≤ 0.01), as well as COL3A1, P4HA2, P4Hβ, and LOXL4 (p ≤ 0.05). In contrast, IGF-II decreases mRNA levels of the collagen degradation enzymes cathepsin (CTS) K, CTSB, and CTSL and protein levels of CTSK (p ≤ 0.05). The SRY-box transcription factor 9 (SOX9) is overexpressed in SSc lung tissues at the mRNA (p ≤ 0.05) and protein (p ≤ 0.01) levels compared to healthy controls. IGF-II induces SOX9 in lung fibroblasts (p ≤ 0.05) via the IGF1R/IR hybrid receptor, and SOX9 regulates TGFβ2 (p ≤ 0.05), TGFβ3 (p ≤ 0.05), COL3A1 (p ≤ 0.01), and P4HA2 (p ≤ 0.001) downstream of IGF-II. Our results identify a novel IGF-II signaling axis and downstream targets that are regulated in a SOX9-dependent and -independent manner. Our findings provide novel insights on the role of IGF-II in promoting pulmonary fibrosis.
Collapse
Affiliation(s)
- Kristy M. Waldrep
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Jessalyn I. Rodgers
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Sara M. Garrett
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| | - Bethany J. Wolf
- Department of Public Health Sciences, Biostatistics and Bioinformatics, Medical University of South Carolina, Charleston, SC 29425, USA;
| | - Carol A. Feghali-Bostwick
- Department of Medicine, Rheumatology, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.W.); (J.I.R.); (S.M.G.)
| |
Collapse
|
14
|
Arif M, Basu A, Wolf KM, Park JK, Pommerolle L, Behee M, Gochuico BR, Cinar R. An Integrative Multiomics Framework for Identification of Therapeutic Targets in Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207454. [PMID: 37038090 PMCID: PMC10238219 DOI: 10.1002/advs.202207454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/26/2023] [Indexed: 06/04/2023]
Abstract
Pulmonary fibrosis (PF) is a heterogeneous disease with a poor prognosis. Therefore, identifying additional therapeutic modalities is required to improve outcome. However, the lack of biomarkers of disease progression hampers the preclinical to clinical translational process. Here, this work assesses and identifies progressive alterations in pulmonary function, transcriptomics, and metabolomics in the mouse lung at 7, 14, 21, and 28 days after a single dose of oropharyngeal bleomycin. By integrating multi-omics data, this work identifies two central gene subnetworks associated with multiple critical pathological changes in transcriptomics and metabolomics as well as pulmonary function. This work presents a multi-omics-based framework to establish a translational link between the bleomycin-induced PF model in mice and human idiopathic pulmonary fibrosis to identify druggable targets and test therapeutic candidates. This work also indicates peripheral cannabinoid receptor 1 (CB1 R) antagonism as a rational therapeutic target for clinical translation in PF. Mouse Lung Fibrosis Atlas can be accessed freely at https://niaaa.nih.gov/mouselungfibrosisatlas.
Collapse
Affiliation(s)
- Muhammad Arif
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
- Laboratory of Cardiovascular Physiology and Tissue InjuryNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Abhishek Basu
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Kaelin M. Wolf
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Joshua K. Park
- Laboratory of Physiologic StudiesNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Lenny Pommerolle
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Madeline Behee
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| | - Bernadette R. Gochuico
- Medical Genetics BranchNational Human Genome Research InstituteNational Institutes of Health (NIH)BethesdaMD20892USA
| | - Resat Cinar
- Section on Fibrotic DisordersNational Institute on Alcohol Abuse and AlcoholismNational Institutes of HealthRockvilleMD20852USA
| |
Collapse
|
15
|
Yang Q, Miao Q, Chen H, Li D, Luo Y, Chiu J, Wang HJ, Chuvanjyan M, Parmacek MS, Shi W. Myocd regulates airway smooth muscle cell remodeling in response to chronic asthmatic injury. J Pathol 2023; 259:331-341. [PMID: 36484734 PMCID: PMC10107741 DOI: 10.1002/path.6044] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 11/13/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Abnormal growth of airway smooth muscle cells is one of the key features in asthmatic airway remodeling, which is associated with asthma severity. The mechanisms underlying inappropriate airway smooth muscle cell growth in asthma remain largely unknown. Myocd has been reported to act as a key transcriptional coactivator in promoting airway-specific smooth muscle development in fetal lungs. Whether Myocd controls airway smooth muscle remodeling in asthma has not been investigated. Mice with lung mesenchyme-specific deletion of Myocd after lung development were generated, and a chronic asthma model was established by sensitizing and challenging the mice with ovalbumin for a prolonged period. Comparison of the asthmatic pathology between the Myocd knockout mice and the wild-type controls revealed that abrogation of Myocd mitigated airway smooth muscle cell hypertrophy and hyperplasia, accompanied by reduced peri-airway inflammation, decreased fibrillar collagen deposition on airway walls, and attenuation of abnormal mucin production in airway epithelial cells. Our study indicates that Myocd is a key transcriptional coactivator involved in asthma airway remodeling. Inhibition of Myocd in asthmatic airways may be an effective approach to breaking the vicious cycle of asthmatic progression, providing a novel strategy in treating severe and persistent asthma. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Qin Yang
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, PR China
| | - Qing Miao
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hui Chen
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Duo Li
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yongfeng Luo
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joanne Chiu
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hong-Jun Wang
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael Chuvanjyan
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael S Parmacek
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Shi
- Department of Surgery, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
16
|
Oruqaj G, Karnati S, Kotarkonda LK, Boateng E, Bartkuhn M, Zhang W, Ruppert C, Günther A, Bartholin L, Shi W, Baumgart-Vogt E. Transforming Growth Factor-β1 Regulates Peroxisomal Genes/Proteins via Smad Signaling in Idiopathic Pulmonary Fibrosis Fibroblasts and Transgenic Mouse Models. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:259-274. [PMID: 36521562 PMCID: PMC10013039 DOI: 10.1016/j.ajpath.2022.11.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 10/10/2022] [Accepted: 11/04/2022] [Indexed: 12/15/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic human disease with persistent destruction of lung parenchyma. Transforming growth factor-β1 (TGF-β1) signaling plays a pivotal role in the initiation and pathogenesis of IPF. As shown herein, TGF-β1 signaling down-regulated not only peroxisome biogenesis but also the metabolism of these organelles in human IPF fibroblasts. In vitro cell culture observations in human fibroblasts and human lung tissue indicated that peroxisomal biogenesis and metabolic proteins were significantly down-regulated in the lung of 1-month-old transgenic mice expressing a constitutively active TGF-β type I receptor kinase (ALK5). The peroxisome biogenesis protein peroxisomal membrane protein Pex13p (PEX13p) as well as the peroxisomal lipid metabolic enzyme peroxisomal acyl-coenzyme A oxidase 1 (ACOX1) and antioxidative enzyme catalase were highly up-regulated in TGF-β type II receptor and Smad3 knockout mice. This study reports a novel mechanism of peroxisome biogenesis and metabolic regulation via TGF-β1-Smad signaling: interaction of the Smad3 transcription factor with the PEX13 gene in chromatin immunoprecipitation-on-chip assay as well as in a bleomycin-induced pulmonary fibrosis model applied to TGF-β type II receptor knockout mice. Taken together, data from this study suggest that TGF-β1 participates in regulation of peroxisomal biogenesis and metabolism via Smad-dependent signaling, opening up novel strategies for the development of therapeutic approaches to inhibit progression of pulmonary fibrosis patients with IPF.
Collapse
Affiliation(s)
- Gani Oruqaj
- Institute for Anatomy and Cell Biology, Medical Cell Biology, and Biomedical Informatics and Systems Medicine, Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Medical Cell Biology, and Biomedical Informatics and Systems Medicine, Giessen, Germany
| | - Lakshmi Kanth Kotarkonda
- Institute for Anatomy and Cell Biology, Medical Cell Biology, and Biomedical Informatics and Systems Medicine, Giessen, Germany
| | - Eistine Boateng
- Institute for Anatomy and Cell Biology, Medical Cell Biology, and Biomedical Informatics and Systems Medicine, Giessen, Germany
| | - Marek Bartkuhn
- Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
| | - Wenming Zhang
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Clemens Ruppert
- Department of Internal Medicine, Medical Clinic II, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Andreas Günther
- Department of Internal Medicine, Medical Clinic II, German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | | | - Wei Shi
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Medical Cell Biology, and Biomedical Informatics and Systems Medicine, Giessen, Germany.
| |
Collapse
|
17
|
Zhang Z, Li H, Wang G, Zhao G, Li C, Cao Y. Thrombospondin-1 and prolyl 4-hydroxylase subunit alpha 3 as potential biomarkers of salivary gland fibrosis. J Dent Sci 2023. [DOI: 10.1016/j.jds.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
|
18
|
Hendy BA, Fertala J, Nicholson T, Abboud JA, Namdari S, Fertala A. Profibrotic behavior of fibroblasts derived from patients that develop posttraumatic shoulder stiffness. Health Sci Rep 2023; 6:e1100. [PMID: 36817629 PMCID: PMC9933492 DOI: 10.1002/hsr2.1100] [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: 07/10/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/18/2023] Open
Abstract
Background and Aims Arthrofibrosis is a severe scarring condition characterized by joint stiffness and pain. Fundamental to developing arthrofibrotic scars is the accelerated production of procollagen I, a precursor of collagen I molecules that form fibrotic deposits in affected joints. The procollagen I production mechanism comprises numerous elements, including enzymes, protein chaperones, and growth factors. This study aimed to elucidate the differences in the production of vital elements of this mechanism in surgical patients who developed significant posttraumatic arthrofibrosis and those who did not. Methods We studied a group of patients who underwent shoulder arthroscopic repair of the rotator cuff. Utilizing fibroblasts isolated from the patients' rotator intervals, we analyzed their responses to profibrotic stimulation with transforming growth factor β1 (TGFβ1). We compared TGFβ1-dependent changes in the production of procollagen I. We studied auxiliary proteins, prolyl 4-hydroxylase (P4H), and heat shock protein 47 (HSP47), that control procollagen stability and folding. A group of other proteins involved in excessive scar formation, including connective tissue growth factor (CTGF), α smooth muscle actin (αSMA), and fibronectin, was also analyzed. Results We observed robust TGFβ1-dependent increases in the production of CTGF, HSP47, αSMA, procollagen I, and fibronectin in fibroblasts from both groups of patients. In contrast, TGFβ1-dependent P4H production increased only in the stiff-shoulder-derived fibroblasts. Conclusion Results suggest P4H may serve as an element of a mechanism that modulates the fibrotic response after rotator cuff injury.
Collapse
Affiliation(s)
- Benjamin A. Hendy
- Department of Orthopaedic Surgery, Sidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Rothman Institute of Orthopaedics, Shoulder and Elbow ServiceThomas Jefferson University HospitalPhiladelphiaPennsylvaniaUSA
- Present address:
Sequoia Institute for Surgical ServicesVisaliaCAUSA
| | - Jolanta Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Thema Nicholson
- Rothman Institute of Orthopaedics, Shoulder and Elbow ServiceThomas Jefferson University HospitalPhiladelphiaPennsylvaniaUSA
| | - Joseph A. Abboud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Rothman Institute of Orthopaedics, Shoulder and Elbow ServiceThomas Jefferson University HospitalPhiladelphiaPennsylvaniaUSA
| | - Surena Namdari
- Department of Orthopaedic Surgery, Sidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Rothman Institute of Orthopaedics, Shoulder and Elbow ServiceThomas Jefferson University HospitalPhiladelphiaPennsylvaniaUSA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical CollegeThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
19
|
Time and phenotype-dependent transcriptome analysis in AAV-TGFβ1 and Bleomycin-induced lung fibrosis models. Sci Rep 2022; 12:12190. [PMID: 35842487 PMCID: PMC9288451 DOI: 10.1038/s41598-022-16344-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
We have previously established a novel mouse model of lung fibrosis based on Adeno-associated virus (AAV)-mediated pulmonary overexpression of TGFβ1. Here, we provide an in-depth characterization of phenotypic and transcriptomic changes (mRNA and miRNA) in a head-to-head comparison with Bleomycin-induced lung injury over a 4-week disease course. The analyses delineate the temporal state of model-specific and commonly altered pathways, thereby providing detailed insights into the processes underlying disease development. They further guide appropriate model selection as well as interventional study design. Overall, Bleomycin-induced fibrosis resembles a biphasic process of acute inflammation and subsequent transition into fibrosis (with partial resolution), whereas the TGFβ1-driven model is characterized by pronounced and persistent fibrosis with concomitant inflammation and an equally complex disease phenotype as observed upon Bleomycin instillation. Finally, based on an integrative approach combining lung function data, mRNA/miRNA profiles, their correlation and miRNA target predictions, we identify putative drug targets and miRNAs to be explored as therapeutic candidates for fibrotic diseases. Taken together, we provide a comprehensive analysis and rich data resource based on RNA-sequencing, along with a strategy for transcriptome-phenotype coupling. The results will be of value for TGFβ research, drug discovery and biomarker identification in progressive fibrosing interstitial lung diseases.
Collapse
|
20
|
Niu X, Ren L, Wang S, Gao D, Ma M, Hu A, Qi H, Zhang S. High Prolyl 4-Hydroxylase Subunit Alpha 3 Expression as an Independent Prognostic Biomarker and Correlated With Immune Infiltration in Gastric Cancer. Front Genet 2022; 13:952335. [PMID: 35846138 PMCID: PMC9283575 DOI: 10.3389/fgene.2022.952335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Gastric cancer (GC) has a high mortality rate and is particularly prevalent in China. The extracellular matrix protein, prolyl 4-hydroxylase subunit alpha 3 (P4HA3), has been implicated in various cancers. We aimed to assess the diagnostic and prognostic value of P4HA3 in GC and investigate its correlation with immune cell infiltration. Methods: The present study used microarray data from the Cancer Genome Atlas (TCGA) to analyze the association of P4HA3 expression with clinicopathological features. Data from the Gene Expression Omnibus (GEO) were used for validation. Receiver operating characteristic (ROC) and Kaplan–Meier curves were constructed to determine the diagnostic and prognostic value of P4HA3 in GC. Univariate and multivariate regression analyses were performed to assess the impact of P4HA3 on overall survival (OS) rates. A protein–protein interaction (PPI) network was generated and functional enrichment evaluated. Single-sample gene set enrichment analysis (ssGSEA) was conducted to correlate P4HA3 expression with immune cell infiltration. The correlation between P4HA3 and immune check point genes was studied. Results: P4HA3 was over-expressed in GC, along with 15 other types of cancer, including breast invasive carcinoma and cholangiocarcinoma. P4HA3 showed high diagnostic and prognostic value in GC and was an independent prognostic factor. P4HA3, TNM (tumor, node, metastases) stage, pathological stage and age all correlated with OS rates. Genes related to P4HA3 were enriched in the lumen of the endoplasmic reticulum and included procollagen-proline 3-dioxygenase activity. P4HA3 expression correlated with numbers of macrophages, natural killer (NK) cells, immature dendritic cells (iDC), mast cells, eosinophils, effective memory T cells (Tem), T-helper 1 (Th1) cells and dendritic cells (DC). P4HA3 was positively correlated with hepatitis A virus cellular receptor 2 (HAVCR2) and programmed cell death 1 ligand 2 (PDCD1LG2). Conclusion: P4HA3 is a potential independent biomarker for prognosis of GC and may be an immunotherapy target in the treatment of GC.
Collapse
Affiliation(s)
- Xiaoji Niu
- Department of Gastroenterology of Traditional Chinese Medicine, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liman Ren
- Department of Endocrinology, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
| | - Shoumei Wang
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dong Gao
- Department of Gastroenterology of Traditional Chinese Medicine, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
| | - Mingyue Ma
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Aiyan Hu
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongjun Qi
- Department of Gastroenterology of Traditional Chinese Medicine, Qinghai Province Hospital of Traditional Chinese Medicine, Xining, China
- *Correspondence: Hongjun Qi, ; Shuhui Zhang,
| | - Shuhui Zhang
- Department of Pathology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- *Correspondence: Hongjun Qi, ; Shuhui Zhang,
| |
Collapse
|
21
|
Xu X, Ma C, Wu H, Ma Y, Liu Z, Zhong P, Jin C, Ning W, Wu X, Zhang Y, Han J, Wang J. Fructose Induces Pulmonary Fibrotic Phenotype Through Promoting Epithelial-Mesenchymal Transition Mediated by ROS-Activated Latent TGF-β1. Front Nutr 2022; 9:850689. [PMID: 35711535 PMCID: PMC9197188 DOI: 10.3389/fnut.2022.850689] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/21/2022] [Indexed: 12/12/2022] Open
Abstract
Fructose is a commonly used food additive and has many adverse effects on human health, but it is unclear whether fructose impacts pulmonary fibrosis. TGF-β1, a potent fibrotic inducer, is produced as latent complexes by various cells, including alveolar epithelial cells, macrophages, and fibroblasts, and must be activated by many factors such as reactive oxygen species (ROS). This study explored the impact of fructose on pulmonary fibrotic phenotype and epithelial-mesenchymal transition (EMT) using lung epithelial cells (A549 or BEAS-2B) and the underlying mechanisms. Fructose promoted the cell viability of lung epithelial cells, while N-Acetyl-l-cysteine (NAC) inhibited such. Co-treatment of fructose and latent TGF-β1 could induce the fibrosis phenotype and the epithelial-mesenchymal transition (EMT)-related protein expression, increasing lung epithelial cell migration and invasion. Mechanism analysis shows that fructose dose-dependently promoted the production of total and mitochondrial ROS in A549 cells, while NAC eliminated this promotion. Notably, post-administration with NAC or SB431542 (a potent TGF-β type I receptor inhibitor) inhibited fibrosis phenotype and EMT process of lung epithelial cells co-treated with fructose and latent TGF-β1. Finally, the fibrosis phenotype and EMT-related protein expression of lung epithelial cells were mediated by the ROS-activated latent TGF-β1/Smad3 signal. This study revealed that high fructose promoted the fibrotic phenotype of human lung epithelial cells by up-regulating oxidative stress, which enabled the latent form of TGF-β1 into activated TGF-β1, which provides help and reference for the diet adjustment of healthy people and patients with fibrosis.
Collapse
Affiliation(s)
- Xiaoxiao Xu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Chuang Ma
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Hang Wu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Yuanqiao Ma
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Zejin Liu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Peijie Zhong
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Chaolei Jin
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Wenjuan Ning
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Xiao Wu
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Yijie Zhang
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Jichang Han
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| | - Junpeng Wang
- Infection and Immunity Institute and Translational Medical Center of Huaihe Hospital, Henan University, Kaifeng, China
| |
Collapse
|
22
|
Staab-Weijnitz CA. Fighting the Fiber: Targeting Collagen in Lung Fibrosis. Am J Respir Cell Mol Biol 2021; 66:363-381. [PMID: 34861139 DOI: 10.1165/rcmb.2021-0342tr] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Organ fibrosis is characterized by epithelial injury and aberrant tissue repair, where activated effector cells, mostly fibroblasts and myofibroblasts, excessively deposit collagen into the extracellular matrix. Fibrosis frequently results in organ failure and has been estimated to contribute to at least one third of all global deaths. Also lung fibrosis, in particular idiopathic pulmonary fibrosis (IPF), is a fatal disease with rising incidence worldwide. As current treatment options targeting fibrogenesis are insufficient, there is an urgent need for novel therapeutic strategies. During the last decade, several studies have proposed to target intra- and extracellular components of the collagen biosynthesis, maturation, and degradation machinery. This includes intra- and extracellular targets directly acting on collagen gene products, but also such that anabolize essential building blocks of collagen, in particular glycine and proline biosynthetic enzymes. Collagen, however, is a ubiquitous molecule in the body and fulfils essential functions as a macromolecular scaffold, growth factor reservoir, and receptor binding site in virtually every tissue. This review summarizes recent advances and future directions in this field. Evidence for the proposed therapeutic targets and where they currently stand in terms of clinical drug development for treatment of fibrotic disease is provided. The drug targets are furthermore discussed in light of (1) specificity for collagen biosynthesis, maturation and degradation, and (2) specificity for disease-associated collagen. As therapeutic success and safety of these drugs may largely depend on targeted delivery, different strategies for specific delivery to the main effector cells and to the extracellular matrix are discussed.
Collapse
Affiliation(s)
- Claudia A Staab-Weijnitz
- Helmholtz Zentrum Munchen Deutsches Forschungszentrum fur Gesundheit und Umwelt, 9150, Comprehensive Pneumology Center/Institute of Lung Biology and Disease, Member of the German Center of Lung Research (DZL), München, Germany;
| |
Collapse
|
23
|
Liu W, Gao M, Li L, Chen Y, Fan H, Cai Q, Shi Y, Pan C, Liu J, Cheng LS, Yang H, Cheng G. Homeoprotein SIX1 compromises antitumor immunity through TGF-β-mediated regulation of collagens. Cell Mol Immunol 2021; 18:2660-2672. [PMID: 34782761 PMCID: PMC8633173 DOI: 10.1038/s41423-021-00800-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
The tumor microenvironment (TME), including infiltrated immune cells, is known to play an important role in tumor growth; however, the mechanisms underlying tumor immunogenicity have not been fully elucidated. Here, we discovered an unexpected role for the transcription factor SIX1 in regulating the tumor immune microenvironment. Based on analyses of patient datasets, we found that SIX1 was upregulated in human tumor tissues and that its expression levels were negatively correlated with immune cell infiltration in the TME and the overall survival rates of cancer patients. Deletion of Six1 in cancer cells significantly reduced tumor growth in an immune-dependent manner with enhanced antitumor immunity in the TME. Mechanistically, SIX1 was required for the expression of multiple collagen genes via the TGFBR2-dependent Smad2/3 activation pathway, and collagen deposition in the TME hampered immune cell infiltration and activation. Thus, our study uncovers a crucial role for SIX1 in modulating tumor immunogenicity and provides proof-of-concept evidence for targeting SIX1 in cancer immunotherapy.
Collapse
Affiliation(s)
- Wancheng Liu
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Meiling Gao
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Lili Li
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Yu Chen
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Huimin Fan
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Qiaomei Cai
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Yueyue Shi
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Chaohu Pan
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Junxiao Liu
- grid.506261.60000 0001 0706 7839Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005 China ,grid.494590.5Suzhou Institute of Systems Medicine, Suzhou, 215123 China
| | - Lucy S. Cheng
- grid.412689.00000 0001 0650 7433Department of Dermatology, University of Pittsburgh Medical Center, 3708 Fifth Avenue, Suite 500.68, Pittsburgh, PA 15213 USA
| | - Heng Yang
- Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China. .,Suzhou Institute of Systems Medicine, Suzhou, 215123, China.
| | - Genhong Cheng
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
24
|
Abstract
The fibrocartilage chondrocyte phenotype has been recognized to attribute to osteoarthritis (OA) development. These chondrocytes express genes related to unfavorable OA outcomes, emphasizing its importance in OA pathology. BMP7 is being explored as a potential disease-modifying molecule and attenuates the chondrocyte hypertrophic phenotype. On the other hand, BMP7 has been demonstrated to relieve organ fibrosis by counteracting the pro-fibrotic TGFβ-Smad3-PAI1 axis and increasing MMP2-mediated Collagen type I turnover. Whether BMP7 has anti-fibrotic properties in chondrocytes is unknown. Human OA articular chondrocytes (HACs) were isolated from end-stage OA femoral cartilage (total knee arthroplasty; n = 18 individual donors). SW1353 cells and OA HACs were exposed to 1 nM BMP7 for 24 h, after which gene expression of fibrosis-related genes and fibrosis-mediating factors was determined by RT-qPCR. In SW1353, Collagen type I protein levels were determined by immunocytochemistry and western blotting. PAI1 and MMP2 protein levels and activity were measured with an ELISA and activity assays, respectively. MMP2 activity was inhibited with the selective MMP-2 inhibitor OA-Hy. SMAD3 activity was determined by a (CAGA)12-reporter assay, and pSMAD2 levels by western blotting. Following BMP7 exposure, the expression of fibrosis-related genes was reduced in SW1353 cells and OA HACs. BMP7 reduced Collagen type I protein levels in SW1353 cells. Gene expression of MMP2 was increased in SW1353 cells following BMP7 treatment. BMP7 reduced PAI1 protein levels and -activity, while MMP2 protein levels and -activity were increased by BMP7. BMP7-dependent inhibition of Collagen type I protein levels in SW1353 cells was abrogated when MMP2 activity was inhibited. Finally, BMP7 reduced pSMAD2 levels determined by western blotting and reduced SMAD3 transcriptional activity as demonstrated by decreased (CAGA)12 luciferase reporter activity. Our data demonstrate that short-term exposure to BMP7 decreases the fibrocartilage chondrocyte phenotype. The BMP7-dependent reduction of Collagen type I protein expression seems MMP2-dependent and inhibition of Smad2/3-PAI1 activity was identified as a potential pathway via which BMP7 exerts its anti-fibrotic action. This indicates that in chondrocytes BMP7 may have a double mode-of-action by targeting both the hypertrophic as well as the fibrotic chondrocyte phenotype, potentially adding to the clinical relevance of using BMP7 as an OA disease-modifying molecule.
Collapse
|
25
|
Bernau K, Leet JP, Bruhn EM, Tubbs AJ, Zhu T, Sandbo N. Expression of serum response factor in the lung mesenchyme is essential for development of pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol 2021; 321:L174-L188. [PMID: 33978489 PMCID: PMC8321854 DOI: 10.1152/ajplung.00323.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Extracellular matrix deposition characterizes idiopathic pulmonary fibrosis (IPF) and is orchestrated by myofibroblasts. The lung mesenchyme is an essential source of myofibroblasts in pulmonary fibrosis. Although the transcription factor serum response factor (SRF) has shown to be critical in the process of myofibroblast differentiation, its role in development of pulmonary fibrosis has not been determined in vivo. In this study, we observed that SRF expression localized to mesenchymal compartments, areas of dense fibrosis, and fibroblastic foci in human (IPF and normal) and bleomycin-treated mouse lungs. To determine the role of mesenchymal SRF in pulmonary fibrosis, we utilized a doxycycline-inducible, Tbx4 lung enhancer (Tbx4LE)-driven Cre-recombinase to disrupt SRF expression in the lung mesenchyme in vivo. Doxycycline-treated Tbx4LE-rtTA/TetO-Cre/tdTom/SRFf,f (and controls) were treated with a single intratracheal dose of bleomycin to induce pulmonary fibrosis and examined for lung mesenchymal expansion, pulmonary fibrosis, and inflammatory response. Bleomycin-treated Tbx4LE-rtTA/TetO-Cre/tdTom/SRFf,f mice showed decreased numbers of Tbx4LE-positive lung mesenchymal cells (LMCs) and collagen accumulation (via hydroxyproline assay) compared with controls. This effect was associated with SRF-null LMCs losing their proliferative and myofibroblast differentiation potential compared with SRF-positive controls. Together, these data demonstrate that SRF plays a critical role in LMC myofibroblast expansion during bleomycin-induced pulmonary fibrosis. This sets the stage for pharmacological strategies that specifically target SRF in the lung mesenchyme as a potential means of treating pulmonary fibrosis.
Collapse
Affiliation(s)
- Ksenija Bernau
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jonathan Paul Leet
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ellen Marie Bruhn
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Austin James Tubbs
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Terry Zhu
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Nathan Sandbo
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
26
|
TGF-β-dependent reprogramming of amino acid metabolism induces epithelial-mesenchymal transition in non-small cell lung cancers. Commun Biol 2021; 4:782. [PMID: 34168290 PMCID: PMC8225889 DOI: 10.1038/s42003-021-02323-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 06/08/2021] [Indexed: 01/06/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT)—a fundamental process in embryogenesis and wound healing—promotes tumor metastasis and resistance to chemotherapy. While studies have identified signaling components and transcriptional factors responsible in the TGF-β-dependent EMT, whether and how intracellular metabolism is integrated with EMT remains to be fully elucidated. Here, we showed that TGF-β induces reprogramming of intracellular amino acid metabolism, which is necessary to promote EMT in non-small cell lung cancer cells. Combined metabolome and transcriptome analysis identified prolyl 4-hydroxylase α3 (P4HA3), an enzyme implicated in cancer metabolism, to be upregulated during TGF-β stimulation. Further, knockdown of P4HA3 diminished TGF-β-dependent changes in amino acids, EMT, and tumor metastasis. Conversely, manipulation of extracellular amino acids induced EMT-like responses without TGF-β stimulation. These results suggest a previously unappreciated requirement for the reprogramming of amino acid metabolism via P4HA3 for TGF-β-dependent EMT and implicate a P4HA3 inhibitor as a potential therapeutic agent for cancer. Through metabolome and transcriptome analyses, Nakasuka et al find that TGF-β-induced epithelial–mesenchymal transition (EMT) in non-small cell lung cancer cells is associated with reprogramming of amino acid metabolism. They also identify P4HA3 as a key enzyme involved in these changes altogether providing insights into potential mechanisms of metastasis.
Collapse
|
27
|
Onursal C, Dick E, Angelidis I, Schiller HB, Staab-Weijnitz CA. Collagen Biosynthesis, Processing, and Maturation in Lung Ageing. Front Med (Lausanne) 2021; 8:593874. [PMID: 34095157 PMCID: PMC8172798 DOI: 10.3389/fmed.2021.593874] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 03/24/2021] [Indexed: 12/15/2022] Open
Abstract
In addition to providing a macromolecular scaffold, the extracellular matrix (ECM) is a critical regulator of cell function by virtue of specific physical, biochemical, and mechanical properties. Collagen is the main ECM component and hence plays an essential role in the pathogenesis and progression of chronic lung disease. It is well-established that many chronic lung diseases, e.g., chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) primarily manifest in the elderly, suggesting increased susceptibility of the aged lung or accumulated alterations in lung structure over time that favour disease. Here, we review the main steps of collagen biosynthesis, processing, and turnover and summarise what is currently known about alterations upon lung ageing, including changes in collagen composition, modification, and crosslinking. Recent proteomic data on mouse lung ageing indicates that, while the ER-resident machinery of collagen biosynthesis, modification and triple helix formation appears largely unchanged, there are specific changes in levels of type IV and type VI as well as the two fibril-associated collagens with interrupted triple helices (FACIT), namely type XIV and type XVI collagens. In addition, levels of the extracellular collagen crosslinking enzyme lysyl oxidase are decreased, indicating less enzymatically mediated collagen crosslinking upon ageing. The latter contrasts with the ageing-associated increase in collagen crosslinking by advanced glycation endproducts (AGEs), a result of spontaneous reactions of protein amino groups with reactive carbonyls, e.g., from monosaccharides or reactive dicarbonyls like methylglyoxal. Given the slow turnover of extracellular collagen such modifications accumulate even more in ageing tissues. In summary, the collective evidence points mainly toward age-induced alterations in collagen composition and drastic changes in the molecular nature of collagen crosslinks. Future work addressing the consequences of these changes may provide important clues for prevention of lung disease and for lung bioengineering and ultimately pave the way to novel targeted approaches in lung regenerative medicine.
Collapse
Affiliation(s)
- Ceylan Onursal
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Elisabeth Dick
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Ilias Angelidis
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Herbert B Schiller
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Claudia A Staab-Weijnitz
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz-Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
28
|
Ruan H, Gao S, Li S, Luan J, Jiang Q, Li X, Yin H, Zhou H, Yang C. Deglycosylated Azithromycin Attenuates Bleomycin-Induced Pulmonary Fibrosis via the TGF-β1 Signaling Pathway. Molecules 2021; 26:molecules26092820. [PMID: 34068694 PMCID: PMC8126120 DOI: 10.3390/molecules26092820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive, life-threatening lung disease characterized by the proliferation of myofibroblasts and deposition of extracellular matrix that results in irreversible distortion of the lung structure and the formation of focal fibrosis. The molecular mechanism of IPF is not fully understood, and there is no satisfactory treatment. However, most studies suggest that abnormal activation of transforming growth factor-β1 (TGF-β1) can promote fibroblast activation and epithelial to mesenchymal transition (EMT) to induce pulmonary fibrosis. Deglycosylated azithromycin (Deg-AZM) is a compound we previously obtained by removing glycosyls from azithromycin; it was demonstrated to exert little or no antibacterial effects. Here, we discovered a new function of Deg-AZM in pulmonary fibrosis. In vivo experiments showed that Deg-AZM could significantly reduce bleomycin-induced pulmonary fibrosis and restore respiratory function. Further study revealed the anti-inflammatory and antioxidant effects of Deg-AZM in vivo. In vitro experiments showed that Deg-AZM inhibited TGF-β1 signaling, weakened the activation and differentiation of lung fibroblasts, and inhibited TGF-β1-induced EMT in alveolar epithelial cells. In conclusion, our findings show that Deg-AZM exerts antifibrotic effects by inhibiting TGF-β1-induced myofibroblast activation and EMT.
Collapse
Affiliation(s)
- Hao Ruan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
| | - Shaoyan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
| | - Shuangling Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Jiaoyan Luan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Qiuyan Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
| | - Huijun Yin
- China Resources Pharmaceutical Group Limited, Beijing 100000, China
- Correspondence: (H.Y.); (H.Z.); (C.Y.); Tel.: +1-331-111-3030 (H.Y.); +1-502-220-6769 (H.Z.); +1-590-135-1388 (C.Y.)
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
- Correspondence: (H.Y.); (H.Z.); (C.Y.); Tel.: +1-331-111-3030 (H.Y.); +1-502-220-6769 (H.Z.); +1-590-135-1388 (C.Y.)
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300000, China; (H.R.); (S.G.); (S.L.); (J.L.); (Q.J.); (X.L.)
- High-throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin 300070, China
- Correspondence: (H.Y.); (H.Z.); (C.Y.); Tel.: +1-331-111-3030 (H.Y.); +1-502-220-6769 (H.Z.); +1-590-135-1388 (C.Y.)
| |
Collapse
|
29
|
van Geffen C, Deißler A, Quante M, Renz H, Hartl D, Kolahian S. Regulatory Immune Cells in Idiopathic Pulmonary Fibrosis: Friends or Foes? Front Immunol 2021; 12:663203. [PMID: 33995390 PMCID: PMC8120991 DOI: 10.3389/fimmu.2021.663203] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
The immune system is receiving increasing attention for interstitial lung diseases, as knowledge on its role in fibrosis development and response to therapies is expanding. Uncontrolled immune responses and unbalanced injury-inflammation-repair processes drive the initiation and progression of idiopathic pulmonary fibrosis. The regulatory immune system plays important roles in controlling pathogenic immune responses, regulating inflammation and modulating the transition of inflammation to fibrosis. This review aims to summarize and critically discuss the current knowledge on the potential role of regulatory immune cells, including mesenchymal stromal/stem cells, regulatory T cells, regulatory B cells, macrophages, dendritic cells and myeloid-derived suppressor cells in idiopathic pulmonary fibrosis. Furthermore, we review the emerging role of regulatory immune cells in anti-fibrotic therapy and lung transplantation. A comprehensive understanding of immune regulation could pave the way towards new therapeutic or preventive approaches in idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Chiel van Geffen
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany
| | - Astrid Deißler
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany.,Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Markus Quante
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Marburg, Germany
| | - Dominik Hartl
- Department of Pediatrics I, Eberhard Karls University of Tübingen, Tübingen, Germany.,Dominik Hartl, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Saeed Kolahian
- Department of Experimental and Clinical Pharmacology and Pharmacogenomics, University Hospital Tübingen, Tübingen, Germany.,Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University of Marburg, Marburg, Germany.,Universities of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Marburg, Germany
| |
Collapse
|
30
|
Käsmann L, Dietrich A, Staab-Weijnitz CA, Manapov F, Behr J, Rimner A, Jeremic B, Senan S, De Ruysscher D, Lauber K, Belka C. Radiation-induced lung toxicity - cellular and molecular mechanisms of pathogenesis, management, and literature review. Radiat Oncol 2020; 15:214. [PMID: 32912295 PMCID: PMC7488099 DOI: 10.1186/s13014-020-01654-9] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Lung, breast, and esophageal cancer represent three common malignancies with high incidence and mortality worldwide. The management of these tumors critically relies on radiotherapy as a major part of multi-modality care, and treatment-related toxicities, such as radiation-induced pneumonitis and/or lung fibrosis, are important dose limiting factors with direct impact on patient outcomes and quality of life. In this review, we summarize the current understanding of radiation-induced pneumonitis and pulmonary fibrosis, present predictive factors as well as recent diagnostic and therapeutic advances. Novel candidates for molecularly targeted approaches to prevent and/or treat radiation-induced pneumonitis and pulmonary fibrosis are discussed.
Collapse
Affiliation(s)
- Lukas Käsmann
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Alexander Dietrich
- Walther Straub Institute of Pharmacology and Toxicology, Member of the German Center for Lung Research (DZL), Medical Faculty, LMU-Munich, Munich, Germany
| | - Claudia A. Staab-Weijnitz
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- Institute of Lung Biology and Disease, Helmholtz Zentrum München, Munich, Germany
| | - Farkhad Manapov
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Jürgen Behr
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- Department of Internal Medicine V, LMU Munich, Munich, Germany
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, USA
| | | | - Suresh Senan
- Department of Radiation Oncology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Dirk De Ruysscher
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Kirsten Lauber
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Marchioninistrasse 15, 81377 Munich, Germany
- German Center for Lung Research (DZL), partner site Munich, Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| |
Collapse
|
31
|
Age-associated genes in human mammary gland drive human breast cancer progression. Breast Cancer Res 2020; 22:64. [PMID: 32539762 PMCID: PMC7294649 DOI: 10.1186/s13058-020-01299-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/20/2020] [Indexed: 12/16/2022] Open
Abstract
Background Aging is a comorbidity of breast cancer suggesting that aging-associated transcriptome changes may promote breast cancer progression. However, the mechanism underlying the age effect on breast cancer remains poorly understood. Method We analyzed transcriptomics of the matched normal breast tissues from the 82 breast cancer patients in The Cancer Genome Atlas (TCGA) dataset with linear regression for genes with age-associated expression that are not associated with menopause. We also analyzed differentially expressed genes between the paired tumor and non-tumor breast tissues in TCGA for the identification of age and breast cancer (ABC)-associated genes. A few of these genes were selected for further investigation of their malignancy-regulating activities with in vitro and in vivo assays. Results We identified 148 upregulated and 189 downregulated genes during aging. Overlapping of tumor-associated genes between normal and tumor tissues with age-dependent genes resulted in 14 upregulated and 24 downregulated genes that were both age and breast cancer associated. These genes are predictive in relapse-free survival, indicative of their potential tumor promoting or suppressive functions, respectively. Knockdown of two upregulated genes (DYNLT3 and P4HA3) or overexpression of the downregulated ALX4 significantly reduced breast cancer cell proliferation, migration, and clonogenicity. Moreover, knockdown of P4HA3 reduced growth and metastasis whereas overexpression of ALX4 inhibited the growth of xenografted breast cancer cells in mice. Conclusion Our study suggests that transcriptome alterations during aging may contribute to breast tumorigenesis. DYNLT3, P4HA3, and ALX4 play significant roles in breast cancer progression.
Collapse
|
32
|
Wang J, Xu L, Xiang Z, Ren Y, Zheng X, Zhao Q, Zhou Q, Zhou Y, Xu L, Wang Y. Microcystin-LR ameliorates pulmonary fibrosis via modulating CD206 + M2-like macrophage polarization. Cell Death Dis 2020; 11:136. [PMID: 32075954 PMCID: PMC7031231 DOI: 10.1038/s41419-020-2329-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 02/08/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a group of chronic interstitial pulmonary diseases characterized by myofibroblast proliferation and extracellular matrix deposition with limited treatment options. Based on our previous observation, we hypothesized microcystin-leucine arginine (LR), an environmental cyanobacterial toxin, could potentially suppress pulmonary fibrosis. In this study, we first demonstrated that chronic exposure of microcystin-LR by oral for weeks indeed attenuated the pulmonary fibrosis both on bleomycin-induced rat and fluorescein isothiocyanate-induced mouse models. Our data further indicated that treatment with microcystin-LR substantially reduced TGF-β1/Smad signaling in rat pulmonary tissues. The experiments in vitro found that microcystin-LR was capable of blocking epithelial–mesenchymal transition (EMT) and fibroblast–myofibroblast transition (FMT) through suppressing the differentiation of CD206+ macrophages. Mechanically, microcystin-LR was found to bind to glucose-regulated protein 78 kDa (GRP78) and suppress endoplasmic reticulum unfolded protein response (UPRER) signaling pathways. These events led to the modulation of M2 polarization of macrophages, which eventually contributed to the alleviation of pulmonary fibrosis. Our results revealed a novel mechanism that may account for therapeutic effect of microcystin-LR on IPF.
Collapse
Affiliation(s)
- Jie Wang
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Lizhi Xu
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Zou Xiang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, China
| | - Yan Ren
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Xiufen Zheng
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Qingya Zhao
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China.,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Qunzhi Zhou
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Yuefen Zhou
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China
| | - Lin Xu
- Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yaping Wang
- Department of Medical Genetics, Nanjing University School of Medicine, Nanjing, 210093, China. .,Jiangsu Key Laboratory of Molecular Medicine, Nanjing University School of Medicine, Nanjing, 210093, China.
| |
Collapse
|
33
|
Experimental pulmonary fibrosis was suppressed by microRNA-506 through NF-kappa-mediated apoptosis and inflammation. Cell Tissue Res 2019; 378:255-265. [DOI: 10.1007/s00441-019-03054-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/28/2019] [Indexed: 12/21/2022]
|
34
|
Butzelaar L, Niessen FB, Talhout W, Schooneman DPM, Ulrich MM, Beelen RHJ, Mink van der Molen AB. Different properties of skin of different body sites: The root of keloid formation? Wound Repair Regen 2017; 25:758-766. [PMID: 28846161 DOI: 10.1111/wrr.12574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 07/03/2017] [Indexed: 01/24/2023]
Abstract
The purpose of this study was to examine extracellular matrix composition, vascularization, and immune cell population of skin sites prone to keloid formation. Keloids remain a complex problem, posing esthetical as well as functional difficulties for those affected. These scars tend to develop at anatomic sites of preference. Mechanical properties of skin vary with anatomic location and depend largely on extracellular matrix composition. These differences in extracellular matrix composition, but also vascularization and resident immune cell populations might play a role in the mechanism of keloid formation. To examine this hypothesis, skin samples of several anatomic locations were taken from 24 human donors within zero to 36 hours after they had deceased. Collagen content and cross-links were determined through high-performance liquid chromatography. The expression of several genes, involved in extracellular matrix production and degradation, was measured by means of real-time PCR. (Immuno)histochemistry was performed to detect fibroblasts, collagen, elastin, blood vessels, Langerhans cells, and macrophages. Properties of skin of keloid predilections sites were compared to properties of skin from other locations (nonpredilection sites [NPS]). The results indicated that there are site specific variations in extracellular matrix properties (collagen and cross-links) as well as macrophage numbers. Moreover, predilection sites (PS) for keloid formation contain larger amounts of collagen compared to NPS, but decreased numbers of macrophages, in particular classically activated CD40 positive macrophages. In conclusion, the altered (histological, protein, and genetic) properties of skin of keloid PS may cause a predisposition for and contribute to keloid formation.
Collapse
Affiliation(s)
- Liselotte Butzelaar
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Frank B Niessen
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands
| | - Wendy Talhout
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Dennis P M Schooneman
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | - Magda M Ulrich
- Department of Plastic, Reconstructive and Hand Surgery, VU University Medical Center, Amsterdam, The Netherlands.,Association of Dutch Burn Centers, Beverwijk, The Netherlands
| | - Robert H J Beelen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, The Netherlands
| | | |
Collapse
|
35
|
Jiang J, Zhang Y, Peng K, Wang Q, Hong X, Li H, Fan G, Zhang Z, Gong T, Sun X. Combined delivery of a TGF-β inhibitor and an adenoviral vector expressing interleukin-12 potentiates cancer immunotherapy. Acta Biomater 2017; 61:114-123. [PMID: 28483693 DOI: 10.1016/j.actbio.2017.05.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 04/25/2017] [Accepted: 05/04/2017] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy appears to have a promising future, but it can be thwarted by secretion of immunosuppressive factors, such as transforming growth factor-β (TGF-β), which inhibits local immune responses to tumors. To weaken immune resistance of tumors and simultaneously strengthen immune responses, we developed a multifunctional polymer that could co-deliver hydrophobic TGF-β inhibitor and an adenovirus gene vector to tumor sites. This co-delivery system sustainably released TGF-β inhibitor SB-505124 and effectively transferred the adenovirus vector carrying the interleukin-12 gene. In addition, it significantly delayed growth of B16 melanoma xenografts in mice and increased animal survival. Mechanistic studies showed that this combination therapy enhanced anti-tumor immune response by activating CD4+ and CD8+ T cells, natural killer cells and interferon-γ secretion in the tumor microenvironment. STATEMENT OF SIGNIFICANCE To weaken immune resistance of tumors and simultaneously strengthen tumors' immune responses, we synthesized a structurally simple, low-toxic but functional polymer β-cyclodextrin-PEI to encapsulate a hydrophobic TGF-β inhibitor SB-505124 and to complex adenovirus vectors expressing IL-12. This is the first report demonstrating that combining TGF-β inhibitor with IL-12 could provide effective immunotherapy against melanoma by the sustainable release of SB-505124 and the effectible transduction of IL-12 gene in tumor cells. The rational delivery system presented a comprehensive and valued platform to be a candidate vector for co-delivering hydrophobic small-molecule drugs and therapeutic genes for treating cancer, providing a new approach for cancer immunotherapy.
Collapse
|
36
|
Hertig V, Tardif K, Meus MA, Duquette N, Villeneuve L, Toussaint F, Ledoux J, Calderone A. Nestin expression is upregulated in the fibrotic rat heart and is localized in collagen-expressing mesenchymal cells and interstitial CD31(+)- cells. PLoS One 2017; 12:e0176147. [PMID: 28448522 PMCID: PMC5407835 DOI: 10.1371/journal.pone.0176147] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022] Open
Abstract
Renal and lung fibrosis was characterized by the accumulation of collagen-immunoreactive mesenchymal cells expressing the intermediate filament protein nestin. The present study tested the hypothesis that nestin expression was increased in the hypertrophied/fibrotic left ventricle of suprarenal abdominal aorta constricted adult male Sprague-Dawley rats and induced in ventricular fibroblasts by pro-fibrotic peptide growth factors. Nestin protein levels were upregulated in the pressure-overloaded left ventricle and expression positively correlated with the rise of mean arterial pressure. In sham and pressure-overloaded hearts, nestin immunoreactivity was detected in collagen type I(+)-and CD31(+)-cells identified in the interstitium and perivascular region whereas staining was absent in smooth muscle α-actin(+)-cells. A significantly greater number of collagen type I(+)-cells co-expressing nestin was identified in the left ventricle of pressure-overloaded rats. Moreover, an accumulation of nestin(+)-cells lacking collagen, CD31 and smooth muscle α-actin staining was selectively observed at the adventitial region of predominantly large calibre blood vessels in the hypertrophied/fibrotic left ventricle. Angiotensin II and TGF-β1 stimulation of ventricular fibroblasts increased nestin protein levels via phosphatidylinositol 3-kinase- and protein kinase C/SMAD3-dependent pathways, respectively. CD31/eNOS(+)-rat cardiac microvascular endothelial cells synthesized/secreted collagen type I, expressed prolyl 4-hydroxylase and TGF-β1 induced nestin expression. The selective accumulation of adventitial nestin(+)-cells highlighted a novel feature of large vessel remodelling in the pressure-overloaded heart and increased appearance of collagen type I/nestin(+)-cells may reflect an activated phenotype of ventricular fibroblasts. CD31/collagen/nestin(+)-interstitial cells could represent displaced endothelial cells displaying an unmasked mesenchymal phenotype, albeit contribution to the reactive fibrotic response of the pressure-overloaded heart remains unknown.
Collapse
Affiliation(s)
- Vanessa Hertig
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Kim Tardif
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Marc Andre Meus
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Natacha Duquette
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Louis Villeneuve
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Fanny Toussaint
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
| | - Jonathan Ledoux
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Medicine, Université de Montréal, Québec, Montréal, Canada
| | - Angelino Calderone
- Research Center, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
- Department of Pharmacology & Physiology, Université de Montréal, Québec, Montréal, Canada
- * E-mail:
| |
Collapse
|
37
|
Cong X, Hubmayr RD, Li C, Zhao X. Plasma membrane wounding and repair in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol 2017; 312:L371-L391. [PMID: 28062486 PMCID: PMC5374305 DOI: 10.1152/ajplung.00486.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/05/2017] [Accepted: 01/05/2017] [Indexed: 12/12/2022] Open
Abstract
Various pathophysiological conditions such as surfactant dysfunction, mechanical ventilation, inflammation, pathogen products, environmental exposures, and gastric acid aspiration stress lung cells, and the compromise of plasma membranes occurs as a result. The mechanisms necessary for cells to repair plasma membrane defects have been extensively investigated in the last two decades, and some of these key repair mechanisms are also shown to occur following lung cell injury. Because it was theorized that lung wounding and repair are involved in the pathogenesis of acute respiratory distress syndrome (ARDS) and idiopathic pulmonary fibrosis (IPF), in this review, we summarized the experimental evidence of lung cell injury in these two devastating syndromes and discuss relevant genetic, physical, and biological injury mechanisms, as well as mechanisms used by lung cells for cell survival and membrane repair. Finally, we discuss relevant signaling pathways that may be activated by chronic or repeated lung cell injury as an extension of our cell injury and repair focus in this review. We hope that a holistic view of injurious stimuli relevant for ARDS and IPF could lead to updated experimental models. In addition, parallel discussion of membrane repair mechanisms in lung cells and injury-activated signaling pathways would encourage research to bridge gaps in current knowledge. Indeed, deep understanding of lung cell wounding and repair, and discovery of relevant repair moieties for lung cells, should inspire the development of new therapies that are likely preventive and broadly effective for targeting injurious pulmonary diseases.
Collapse
Affiliation(s)
- Xiaofei Cong
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia
| | - Rolf D Hubmayr
- Emerius, Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota; and
| | - Changgong Li
- Department of Pediatrics, University of Southern California, Los Angeles, California
| | - Xiaoli Zhao
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, Virginia;
| |
Collapse
|
38
|
Wirsdörfer F, Jendrossek V. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Front Immunol 2016; 7:591. [PMID: 28018357 PMCID: PMC5155013 DOI: 10.3389/fimmu.2016.00591] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Radiation-induced pneumonitis and fibrosis are dose-limiting side effects of thoracic irradiation. Thoracic irradiation triggers acute and chronic environmental lung changes that are shaped by the damage response of resident cells, by the resulting reaction of the immune system, and by repair processes. Although considerable progress has been made during the last decade in defining involved effector cells and soluble mediators, the network of pathophysiological events and the cellular cross talk linking acute tissue damage to chronic inflammation and fibrosis still require further definition. Infiltration of cells from the innate and adaptive immune systems is a common response of normal tissues to ionizing radiation. Herein, lymphocytes represent a versatile and wide-ranged group of cells of the immune system that can react under specific conditions in various ways and participate in modulating the lung environment by adopting pro-inflammatory, anti-inflammatory, or even pro- or anti-fibrotic phenotypes. The present review provides an overview on published data about the role of lymphocytes in radiation-induced lung disease and related damage-associated pulmonary diseases with a focus on T lymphocytes and B lymphocytes. We also discuss the suspected dual role of specific lymphocyte subsets during the pneumonitic phase and fibrotic phase that is shaped by the environmental conditions as well as the interaction and the intercellular cross talk between cells from the innate and adaptive immune systems and (damaged) resident epithelial cells and stromal cells (e.g., endothelial cells, mesenchymal stem cells, and fibroblasts). Finally, we highlight potential therapeutic targets suited to counteract pathological lymphocyte responses to prevent or treat radiation-induced lung disease.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| |
Collapse
|
39
|
Fang J, Wang W, Sun S, Wang Y, Li Q, Lu X, Qiu M, Zhang Y. Metabolomics study of renal fibrosis and intervention effects of total aglycone extracts of Scutellaria baicalensis in unilateral ureteral obstruction rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:20-29. [PMID: 27286917 DOI: 10.1016/j.jep.2016.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/09/2016] [Accepted: 06/04/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellariae Radix (Scutellaria baicalensis Georgi) is a well-known Traditional Chinese Medicine (TCM) which mainly contains flavonoids. Our previous studies have demonstrated that total aglycone extracts of Scutellaria baicalensis (TAES) can improve kidney disease in rats. AIM OF THE STUDY To investigate the renal fibrosis (RF) pathogenesis and TAES treatment mechanism in unilateral ureteral obstruction (UUO) rats, using a metabolomics approach based on gas chromatography-mass spectrometry (GC/MS). METHODS Rats with RF were divided into 6 groups with rats subjected to sham operation as normal control. The effects of TAES on some RF closely related parameters in UUO rats were investigated. A metabolomics method, based on GC/MS, was developed to monitor metabolic alterations in urine. Multivariate data analysis was utilized to identify biomarkers potentially associated with RF and the anti-RF activity of TAES. Ontology-based enrichment analysis by BiNChE and pathway analysis by MetPA aid in the interpretation of difference metabolites. RESULTS After 10 days of treatment, the parameters of renal function begin returning to normal, and the abnormal high expressions of genes associated with extracellular matrix (ECM) were relived. In the metabolomics study, metabolic perturbations induced by UUO were reversed after treatment and TAES showed a dose-dependent therapy effect on RF, meanwhile, 18 potential biomarkers associated with RF were identified. Enrichment analysis of metabolites shows an over representation of mostly alkane-alpha, omega-diamine and alpha, omega-dicarboxylic acid, and these biomarkers are primarily involved in Glycine, serine and threonine metabolism, Retinol metabolism, Arginine and proline metabolism and Fructose and mannose metabolism. CONCLUSIONS Our findings indicate that TAES have positive effects on UUO-induced RF in rats, meanwhile, metabolomics method coupled with metabolites enrichment analysis is a useful tool for revealing the pathogenesis of diseases and action mechanism of TCM on the whole body.
Collapse
Affiliation(s)
- Junwei Fang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenyu Wang
- MacroStat (China) Clinical Research Co., Ltd, Shanghai 201203, China
| | - Shujun Sun
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Wang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qianhua Li
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiong Lu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yongyu Zhang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
40
|
Winslow S, Lindquist KE, Edsjö A, Larsson C. The expression pattern of matrix-producing tumor stroma is of prognostic importance in breast cancer. BMC Cancer 2016; 16:841. [PMID: 27809802 PMCID: PMC5095990 DOI: 10.1186/s12885-016-2864-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/12/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are several indications that the composition of the tumor stroma can contribute to the malignancy of a tumor. Here we utilized expression data sets to identify metagenes that may serve as surrogate marker for the extent of matrix production and vascularization of a tumor and to characterize prognostic molecular components of the stroma. METHODS TCGA data sets from six cancer forms, two breast cancer microarray sets and one mRNA data set of xenografted tumors were downloaded. Using the mean correlation as distance measure compact clusters with genes representing extracellular matrix production (ECM metagene) and vascularization (endothelial metagene) were defined. Explorative Cox modeling was used to identify prognostic stromal gene sets. RESULTS Clustering of stromal genes in six cancer data sets resulted in metagenes, each containing three genes, representing matrix production and vascularization. The ECM metagene was associated with poor prognosis in renal clear cell carcinoma and in lung adenocarcinoma but not in other cancers investigated. Explorative Cox modeling using gene pairs identified gene sets that in multivariate models were prognostic in breast cancer. This was validated in two microarray sets. Two notable genes are TCF4 and P4HA3 which were included in the sets associated with positive and negative prognosis, respectively. Data from laser-microdissected tumors, a xenografted tumor data set and from correlation analyses demonstrate the stroma specificity of the genes. CONCLUSIONS It is possible to construct ECM and endothelial metagenes common for several cancer forms. The molecular composition of matrix-producing cells, rather than the extent of matrix production seem to be important for breast cancer prognosis.
Collapse
Affiliation(s)
- Sofia Winslow
- Department of Laboratory Medicine, Lund University Cancer Center, Translational Cancer Research, Lund University, Lund, Sweden.,Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Kajsa Ericson Lindquist
- Department of Pathology, Regional Laboratories Region Skåne, Lund, Sweden.,Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Anders Edsjö
- Department of Pathology, Regional Laboratories Region Skåne, Lund, Sweden.,Department of Pathology, Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christer Larsson
- Department of Laboratory Medicine, Lund University Cancer Center, Translational Cancer Research, Lund University, Lund, Sweden.
| |
Collapse
|
41
|
Swonger JM, Liu JS, Ivey MJ, Tallquist MD. Genetic tools for identifying and manipulating fibroblasts in the mouse. Differentiation 2016; 92:66-83. [PMID: 27342817 PMCID: PMC5079827 DOI: 10.1016/j.diff.2016.05.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2015] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 01/18/2023]
Abstract
The use of mouse genetic tools to track and manipulate fibroblasts has provided invaluable in vivo information regarding the activities of these cells. Recently, many new mouse strains have been described for the specific purpose of studying fibroblast behavior. Colorimetric reporter mice and lines expressing Cre are available for the study of fibroblasts in the organs prone to fibrosis, including heart, kidney, liver, lung, and skeletal muscle. In this review we summarize the current state of the models that have been used to define tissue resident fibroblast populations. While these complex genetic reagents provide unique insights into the process of fibrosis, they also require a thorough understanding of the caveats and limitations. Here, we discuss the specificity and efficiency of the available genetic models and briefly describe how they have been used to document the mechanisms of fibrosis.
Collapse
Affiliation(s)
- Jessica M Swonger
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Jocelyn S Liu
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Malina J Ivey
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA
| | - Michelle D Tallquist
- Departments of Medicine and Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96813, USA.
| |
Collapse
|
42
|
Wu Q, Han L, Yan W, Ji X, Han R, Yang J, Yuan J, Ni C. miR-489 inhibits silica-induced pulmonary fibrosis by targeting MyD88 and Smad3 and is negatively regulated by lncRNA CHRF. Sci Rep 2016; 6:30921. [PMID: 27506999 PMCID: PMC4978961 DOI: 10.1038/srep30921] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 07/11/2016] [Indexed: 12/22/2022] Open
Abstract
Silicosis is an incurable occupational disease associated with inflammation, fibroblast proliferation and the accumulation of extracellular matrix in lung tissues. The dysregulation of lncRNAs and miRNAs has been implicated in many complex diseases; however, the current understanding of their roles in fibrotic lung diseases, especially silicosis, remains limited. Our previous microRNA (miRNA, miR) microarray data have indicated decreased expression levels of miR-489 in lung tissues of silica-induced pulmonary fibrosis. Here, we further explored the role of miR-489 in a mouse model of silicosis. Interestingly, miR-489 levels were reduced in both macrophages that were exposed to silica and fibroblasts that were exposed to TGF-β1. Additionally, the overexpressed miR-489 carried out its anti-fibrotic role by attenuating inflammation and fibrotic progression in vivo. Our molecular study further demonstrated that miR-489 inhibited silica-induced pulmonary fibrosis primarily by repressing its target genes MyD88 and Smad3. Moreover, the up-regulated lncRNA cardiac hypertrophy-related factor (CHRF) reversed the inhibitory effect of miR-489 on MyD88 and Smad3 and then triggered the inflammation and fibrotic signaling pathways. Overall, our data indicate that the CHRF-miR-489-MyD88 Smad3 signaling axis exerts key functions in silica-induced pulmonary fibrosis and may represent a therapeutic target for silicosis.
Collapse
Affiliation(s)
- Qiuyun Wu
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lei Han
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.,Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, China
| | - Weiwen Yan
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaoming Ji
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ruhui Han
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jingjin Yang
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jiali Yuan
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chunhui Ni
- Department of Occupational Medicine and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
43
|
Xie T, Liang J, Liu N, Huan C, Zhang Y, Liu W, Kumar M, Xiao R, D'Armiento J, Metzger D, Chambon P, Papaioannou VE, Stripp BR, Jiang D, Noble PW. Transcription factor TBX4 regulates myofibroblast accumulation and lung fibrosis. J Clin Invest 2016; 126:3063-79. [PMID: 27400124 DOI: 10.1172/jci85328] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 05/12/2016] [Indexed: 01/21/2023] Open
Abstract
Progressive tissue fibrosis is a major cause of the morbidity and mortality associated with repeated epithelial injuries and accumulation of myofibroblasts. Successful treatment options are limited by an incomplete understanding of the molecular mechanisms that regulate myofibroblast accumulation. Here, we employed in vivo lineage tracing and real-time gene expression transgenic reporting methods to analyze the early embryonic transcription factor T-box gene 4 (TBX4), and determined that TBX4-lineage mesenchymal progenitors are the predominant source of myofibroblasts in injured adult lung. In a murine model, ablation of TBX4-expressing cells or disruption of TBX4 signaling attenuated lung fibrosis after bleomycin-induced injury. Furthermore, TBX4 regulated hyaluronan synthase 2 production to enable fibroblast invasion of matrix both in murine models and in fibroblasts from patients with severe pulmonary fibrosis. These data identify TBX4 as a mesenchymal transcription factor that drives accumulation of myofibroblasts and the development of lung fibrosis. Targeting TBX4 and downstream factors that regulate fibroblast invasiveness could lead to therapeutic approaches in lung fibrosis.
Collapse
|
44
|
Luo Y, Chen H, Ren S, Li N, Mishina Y, Shi W. BMP signaling is essential in neonatal surfactant production during respiratory adaptation. Am J Physiol Lung Cell Mol Physiol 2016; 311:L29-38. [PMID: 27190064 DOI: 10.1152/ajplung.00391.2015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/17/2016] [Indexed: 01/08/2023] Open
Abstract
Deficiency in pulmonary surfactant results in neonatal respiratory distress, and the known genetic mutations in key components of surfactant only account for a small number of cases. Therefore, determining the regulatory mechanisms of surfactant production and secretion, particularly during the transition from prenatal to neonatal stages, is essential for better understanding of the pathogenesis of human neonatal respiratory distress. We have observed significant increase of bone morphogenetic protein (BMP) signaling in neonatal mouse lungs immediately after birth. Using genetically manipulated mice, we then studied the relationship between BMP signaling and surfactant production in neonates. Blockade of endogenous BMP signaling by deleting Bmpr1a (Alk3) or Smad1 in embryonic day 18.5 in perinatal lung epithelial cells resulted in severe neonatal respiratory distress and death, accompanied by atelectasis in histopathology and significant reductions of surfactant protein B and C, as well as Abca3, whereas prenatal lung development was not significantly affected. We then identified a new BMP-Smad1 downstream target, Nfatc3, which is known as an important transcription activator for surfactant proteins and Abca3. Furthermore, activation of BMP signaling in cultured lung epithelial cells was able to promote endogenous Nfatc3 expression and also stimulate the activity of an Nfatc3 promoter that contains a Smad1-binding site. Therefore, our study suggests that the BMP-Alk3-Smad1-Nfatc3 regulatory loop plays an important role in enhancing surfactant production in neonates, possibly helping neonatal respiratory adaptation from prenatal amniotic fluid environment to neonatal air breathing.
Collapse
Affiliation(s)
- Yongfeng Luo
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Hui Chen
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Siying Ren
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Respiratory Medicine, The Second Xiangya Hospital, Central-South University, Changsha, Hunan, People's Republic of China
| | - Nan Li
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Yuji Mishina
- Department of Biologic and Material Sciences, University of Michigan, Ann Arbor, Michigan
| | - Wei Shi
- Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, California; Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California;
| |
Collapse
|
45
|
Kai F, Laklai H, Weaver VM. Force Matters: Biomechanical Regulation of Cell Invasion and Migration in Disease. Trends Cell Biol 2016; 26:486-497. [PMID: 27056543 DOI: 10.1016/j.tcb.2016.03.007] [Citation(s) in RCA: 184] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 03/15/2016] [Accepted: 03/17/2016] [Indexed: 01/05/2023]
Abstract
Atherosclerosis, cancer, and various chronic fibrotic conditions are characterized by an increase in the migratory behavior of resident cells and the enhanced invasion of assorted exogenous cells across a stiffened extracellular matrix (ECM). This stiffened scaffold aberrantly engages cellular mechanosignaling networks in cells, which promotes the assembly of invadosomes and lamellae for cell invasion and migration. Accordingly, deciphering the conserved molecular mechanisms whereby matrix stiffness fosters invadosome and lamella formation could identify therapeutic targets to treat fibrotic conditions, and reducing ECM stiffness could ameliorate disease progression.
Collapse
Affiliation(s)
- FuiBoon Kai
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Hanane Laklai
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA; Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA; Helen Diller Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
46
|
Jiang J, Zhang Y, Zhang Q, Li Y, Gong T, Zhang Z, Ding R, Sun X. Development and validation of an LC–MS/MS method for the determination of SB-505124 in rat plasma: Application to pharmacokinetic study. J Pharm Biomed Anal 2016; 117:205-9. [DOI: 10.1016/j.jpba.2015.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/23/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022]
|