1
|
Zhao Y, Shen L, Yan R, Liu L, Guo P, Liu S, Chen Y, Yuan Z, Gong W, Ji J. Identification of Candidate Lung Function-Related Plasma Proteins to Pinpoint Drug Targets for Common Pulmonary Diseases: A Comprehensive Multi-Omics Integration Analysis. Curr Issues Mol Biol 2025; 47:167. [PMID: 40136421 PMCID: PMC11941423 DOI: 10.3390/cimb47030167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
The genome-wide association studies (GWAS) of lung disease and lung function indices suffer from challenges to be transformed into clinical interventions, due to a lack of knowledge on the molecular mechanism underlying the GWAS associations. A proteome-wide association study (PWAS) was first performed to identify candidate proteins by integrating two independent largest protein quantitative trait loci datasets of plasma proteins and four large-scale GWAS summary statistics of lung function indices (forced expiratory volume in 1 s (FEV1), forced vital capacity (FVC), FEV1/FVC and peak expiratory flow (PEF)), followed by enrichment analysis to reveal the underlying biological processes and pathways. Then, with a discovery dataset, we conducted Mendelian randomization (MR) and Bayesian colocalization analyses to select potentially causal proteins, followed by a replicated MR analysis with an independent dataset. Mediation analysis was also performed to explore the possible mediating role of these indices on the association between proteins and two common lung diseases (chronic obstructive pulmonary disease, COPD and Asthma). We finally prioritized the potential drug targets. A total of 210 protein-lung function index associations were identified by PWAS, and were significantly enriched in the pulmonary fibrosis and lung tissue repair. Subsequent MR and colocalization analysis identified 59 causal protein-index pairs, among which 42 pairs were replicated. Further mediation analysis identified 3 potential pathways from proteins to COPD or asthma mediated by FEV1/FVC. The mediated proportion ranges from 68.4% to 82.7%. Notably, 24 proteins were reported as druggable targets in Drug Gene Interaction Database, among which 8 were reported to interact with drugs, including FKBP4, GM2A, COL6A3, MAPK3, SERPING1, XPNPEP1, DNER, and FER. Our study identified the crucial plasma proteins causally associated with lung functions and highlighted potential mediating mechanism underlying the effect of proteins on common lung diseases. These findings may have an important insight into pathogenesis and possible future therapies of lung disorders.
Collapse
Affiliation(s)
- Yansong Zhao
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Lujia Shen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Ran Yan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Lu Liu
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA;
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Ping Guo
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Shuai Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Yingxuan Chen
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Weiming Gong
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Wenhua West Road, Jinan 250012, China; (Y.Z.); (L.S.); (R.Y.); (P.G.); (S.L.); (Y.C.); (Z.Y.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
| | - Jiadong Ji
- Institute for Medical Dataology, Shandong University, 12550, Erhuan East Road, Jinan 250003, China
- Department of Statistics, School of Mathematics, Shandong University, Shanda South Street, Jinan 250100, China
| |
Collapse
|
2
|
Shen W, Wei W, Wang S, Yang X, Wang R, Tian H. RNA-binding protein AZGP1 inhibits epithelial cell proliferation by regulating the genes of alternative splicing in COPD. Gene 2024; 927:148736. [PMID: 38950687 DOI: 10.1016/j.gene.2024.148736] [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: 01/09/2024] [Revised: 05/22/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
BACKGROUND Chronic Obstructive Pulmonary Disease (COPD) is characterized by high morbidity, disability, and mortality rates worldwide. RNA-binding proteins (RBPs) might regulate genes involved in oxidative stress and inflammation in COPD patients. Single-cell transcriptome sequencing (scRNA-seq) offers an accurate tool for identifying intercellular heterogeneity and the diversity of immune cells. However, the role of RBPs in the regulation of various cells, especially AT2 cells, remains elusive. MATERIALS AND METHODS A scRNA-seq dataset (GSE173896) and a bulk RNA-seq dataset acquired from airway tissues (GSE124180) were employed for data mining. Next, RNA-seq analysis was performed in both COPD and control patients. Differentially expressed genes (DEGs) were identified using criteria of fold change (FC ≥ 1.5 or ≤ 1.5) and P value ≤ 0.05. Lastly, Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and alternative splicing identification analyses were carried out. RESULTS RBP genes exhibited specific expression patterns across different cell groups and participated in cell proliferation and mitochondrial dysfunction in AT2 cells. As an RBP, AZGP1 expression was upregulated in both the scRNA-seq and RNA-seq datasets. It might potentially be a candidate immune biomarker that regulates COPD progression by modulating AT2 cell proliferation and adhesion by regulating the expression of SAMD5, DNER, DPYSL3, GBP5, GBP3, and KCNJ2. Moreover, AZGP1 regulated alternative splicing events in COPD, particularly DDAH1 and SFRP1, holding significant implications in COPD. CONCLUSION RBP gene AZGP1 inhibits epithelial cell proliferation by regulating genes participating in alternative splicing in COPD.
Collapse
Affiliation(s)
- Wen Shen
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China.
| | - Wei Wei
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China
| | - Shukun Wang
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China
| | - Xiaolei Yang
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China
| | - Ruili Wang
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China
| | - Hong Tian
- General Medicine Department, The Second Affiliated Hospital of Kunming Medical University, China
| |
Collapse
|
3
|
Fang S, Li Z, Pang S, Gan Y, Ding X, Peng H. Identification of postnatal development dependent genes and proteins in porcine epididymis. BMC Genomics 2023; 24:729. [PMID: 38049726 PMCID: PMC10694963 DOI: 10.1186/s12864-023-09827-y] [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: 04/13/2023] [Accepted: 11/22/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND The epididymis is a highly regionalized tubular organ possesses vectorial functions of sperm concentration, maturation, transport, and storage. The epididymis-expressed genes and proteins are characterized by regional and developmental dependent pattern. However, a systematic and comprehensive insight into the postnatal development dependent changes in gene and protein expressions of porcine epididymis is still lacking. Here, the RNA and protein of epididymis of Duroc pigs at different postnatal development stages were extracted by using commercial RNeasy Midi kit and extraction buffer (7 M Urea, 2 M thiourea, 3% CHAPS, and 1 mM PMSF) combined with sonication, respectively, which were further subjected to transcriptomic and proteomic profiling. RESULTS Transcriptome analysis indicated that 198 and 163 differentially expressed genes (DEGs) were continuously up-regulated and down-regulated along with postnatal development stage changes, respectively. Most of the up-regulated DEGs linked to functions of endoplasmic reticulum and lysosome, while the down-regulated DEGs mainly related to molecular process of extracellular matrix. Moreover, the following key genes INSIG1, PGRMC1, NPC2, GBA, MMP2, MMP14, SFRP1, ELN, WNT-2, COL3A1, and SPARC were highlighted. A total of 49 differentially expressed proteins (DEPs) corresponding to postnatal development stages changes were uncovered by the proteome analysis. Several key proteins ACSL3 and ACADM, VDAC1 and VDAC2, and KNG1, SERPINB1, C3, and TF implicated in fatty acid metabolism, voltage-gated ion channel assembly, and apoptotic and immune processes were emphasized. In the integrative network, the key genes and proteins formed different clusters and showed strong interactions. Additionally, NPC2, COL3A1, C3, and VDAC1 are located at the hub position in each cluster. CONCLUSIONS The identified postnatal development dependent genes and proteins in the present study will pave the way for shedding light on the molecular basis of porcine epididymis functions and are useful for further studies on the specific regulation mechanisms responsible for epididymal sperm maturation.
Collapse
Affiliation(s)
- Shaoming Fang
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 35002, China
| | - Zhechen Li
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 35002, China
| | - Shuo Pang
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 35002, China
| | - Yating Gan
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 35002, China
| | - Xiaoning Ding
- College of Animal Science (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, 35002, China
| | - Hui Peng
- College of Animal Science and Technology, Hainan University, Haikou, 570228, China.
| |
Collapse
|
4
|
Tung VSK, Mathews F, Boruk M, Suppa G, Foronjy R, Pato MT, Pato CN, Knowles JA, Evgrafov OV. Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology. Int J Mol Sci 2023; 24:15339. [PMID: 37895019 PMCID: PMC10607243 DOI: 10.3390/ijms242015339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The study of neurodevelopmental molecular mechanisms in schizophrenia requires the development of adequate biological models such as patient-derived cells and their derivatives. We previously utilized cell lines with neural progenitor properties (CNON) derived from the superior or middle turbinates of patients with schizophrenia and control groups to study schizophrenia-specific gene expression. In this study, we analyzed single-cell RNA seq data from two CNON cell lines (one derived from an individual with schizophrenia (SCZ) and the other from a control group) and two biopsy samples from the middle turbinate (MT) (also from an individual with SCZ and a control). We compared our data with previously published data regarding the olfactory neuroepithelium and demonstrated that CNON originated from a single cell type present both in middle turbinate and the olfactory neuroepithelium and expressed in multiple markers of mesenchymal cells. To define the relatedness of CNON to the developing human brain, we also compared CNON datasets with scRNA-seq data derived from an embryonic brain and found that the expression profile of the CNON closely matched the expression profile one of the cell types in the embryonic brain. Finally, we evaluated the differences between SCZ and control samples to assess the utility and potential benefits of using CNON single-cell RNA seq to study the etiology of schizophrenia.
Collapse
Affiliation(s)
- Victoria Sook Keng Tung
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Fasil Mathews
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Marina Boruk
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Gabrielle Suppa
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Robert Foronjy
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
| | - Michele T. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - Carlos N. Pato
- Department of Psychiatry, Rutgers University, Piscataway, NJ 08854, USA (C.N.P.)
| | - James A. Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| | - Oleg V. Evgrafov
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY 11203, USA
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA;
| |
Collapse
|
5
|
Weckerle J, Mayr CH, Fundel-Clemens K, Lämmle B, Boryn L, Thomas MJ, Bretschneider T, Luippold AH, Huber HJ, Viollet C, Rist W, Veyel D, Ramirez F, Klee S, Kästle M. Transcriptomic and Proteomic Changes Driving Pulmonary Fibrosis Resolution in Young and Old Mice. Am J Respir Cell Mol Biol 2023; 69:422-440. [PMID: 37411041 DOI: 10.1165/rcmb.2023-0012oc] [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: 01/09/2023] [Accepted: 07/06/2023] [Indexed: 07/08/2023] Open
Abstract
Bleomycin-induced pulmonary fibrosis in mice mimics major hallmarks of idiopathic pulmonary fibrosis. Yet in this model, it spontaneously resolves over time. We studied molecular mechanisms of fibrosis resolution and lung repair, focusing on transcriptional and proteomic signatures and the effect of aging. Old mice showed incomplete and delayed lung function recovery 8 weeks after bleomycin instillation. This shift in structural and functional repair in old bleomycin-treated mice was reflected in a temporal shift in gene and protein expression. We reveal gene signatures and signaling pathways that underpin the lung repair process. Importantly, the downregulation of WNT, BMP, and TGFβ antagonists Frzb, Sfrp1, Dkk2, Grem1, Fst, Fstl1, and Inhba correlated with lung function improvement. Those genes constitute a network with functions in stem cell pathways, wound, and pulmonary healing. We suggest that insufficient and delayed downregulation of those antagonists during fibrosis resolution in old mice explains the impaired regenerative outcome. Together, we identified signaling pathway molecules with relevance to lung regeneration that should be tested in-depth experimentally as potential therapeutic targets for pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | - Bärbel Lämmle
- Global Computational Biology and Digital Sciences, and
| | | | | | - Tom Bretschneider
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Andreas H Luippold
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | | | | | - Wolfgang Rist
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Daniel Veyel
- Department of Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany; and
| | - Fidel Ramirez
- Global Computational Biology and Digital Sciences, and
| | - Stephan Klee
- Department of Immunology and Respiratory Disease Research
| | - Marc Kästle
- Department of Immunology and Respiratory Disease Research
| |
Collapse
|
6
|
Tung VSK, Mathews F, Boruk M, Suppa G, Foronjy R, Pato M, Pato C, Knowles JA, Evgrafov OV. Cultured Mesenchymal Cells from Nasal Turbinate as a Cellular Model of the Neurodevelopmental Component of Schizophrenia Etiology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.28.534295. [PMID: 37034711 PMCID: PMC10081251 DOI: 10.1101/2023.03.28.534295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Study of the neurodevelopmental molecular mechanisms of schizophrenia requires the development of adequate biological models such as patient-derived cells and their derivatives. We previously used cell lines with neural progenitor properties (CNON) derived from superior or middle turbinates of patients with schizophrenia and control groups to study gene expression specific to schizophrenia. In this study, we compared single cell-RNA seq data from two CNON cell lines, one derived from an individual with schizophrenia (SCZ) and the other from a control group, with two biopsy samples from the middle turbinate (MT), also from an individual with SCZ and a control. In addition, we compared our data with previously published data from olfactory neuroepithelium (1). Our data demonstrated that CNON originated from a single cell type which is present both in middle turbinate and olfactory neuroepithelium. CNON express multiple markers of mesenchymal cells. In order to define relatedness of CNON to the developing human brain, we also compared CNON datasets with scRNA-seq data of embryonic brain (2) and found that the expression profile of CNON very closely matched one of the cell types in the embryonic brain. Finally, we evaluated differences between SCZ and control samples to assess usability and potential benefits of using single cell RNA-seq of CNON to study etiology of schizophrenia.
Collapse
Affiliation(s)
| | - Fasil Mathews
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Marina Boruk
- Department of Otolaryngology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Gabrielle Suppa
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| | - Robert Foronjy
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| | | | - Carlos Pato
- Department of Psychiatry, Rutgers University
| | - James A. Knowles
- Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ, USA
| | - Oleg V. Evgrafov
- Department of Cell Biology, State University of New York at Downstate, Brooklyn, NY, USA
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW To provide an update on the current understanding of the role of wingless/integrase-1 (Wnt) signaling in pediatric allergic asthma and other pediatric lung diseases. RECENT FINDINGS The Wnt signaling pathway is critical for normal lung development. Genetic and epigenetic human studies indicate a link between Wnt signaling and the development and severity of asthma in children. Mechanistic studies using animal models of allergic asthma demonstrate a key role for Wnt signaling in allergic airway inflammation and remodeling. More recently, data on bronchopulmonary dysplasia (BPD) pathogenesis points to the Wnt signaling pathway as an important regulator. SUMMARY Current data indicates that the Wnt signaling pathway is an important mediator in allergic asthma and BPD pathogenesis. Further studies are needed to characterize the roles of individual Wnt signals in childhood disease, and to identify potential novel therapeutic targets to slow or prevent disease processes.
Collapse
Affiliation(s)
- Nooralam Rai
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Jeanine D’Armiento
- Department of Anesthesiology, Medicine, and Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Predictive Value of Serum Markers SFRP1 and CC16 in Acute Exacerbation of Chronic Obstructive Pulmonary Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6488935. [PMID: 35958937 PMCID: PMC9363185 DOI: 10.1155/2022/6488935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
Abstract
Background Chronic obstructive pulmonary disease (COPD) patients are particularly vulnerable to acute exacerbation, resulting in a huge socioeconomic burden. Objective In this study, we evaluated the value of serum secreted frizzled-related protein 1 (SFRP1) and Clara cell-secreted protein (CC16) in predicting the risk of acute exacerbations in patients with COPD. Methods The study included 123 COPD patients admitted to our hospital from May 2020 to June 2021, including 65 patients in stable stage (STCOPD group), 58 patients in acute exacerbation stage (AECOPD group), and 60 healthy volunteers (control group). Serum SFRP1 and CC16 levels were detected by enzyme-linked immunosorbent assay (ELISA). The receiver operating characteristics curve (ROC) analysis was performed to evaluate the sensitivity and specificity of serum SFRP1 and CC16 for predicting the risk of acute exacerbation in COPD patients. Results The age among groups is significantly different, but there is no difference in the gender and body mass index (BMI). The level of serum SFRP1 in the AECOPD group was significantly higher than that in the STCOPD group and the control group, and the level of serum CC16 was lower than that in the STCOPD group and the control group. Serum SFRP1 was negatively correlated with forced expiratory volume in one second (FEV1)/forced vital capacity (FVC) (r = −0.473, P < 0.001). Serum CC16 was positively correlated with FEV1/FVC (r = 0.457, P < 0.001). The area under the curve (AUC), sensitivity, and specificity of SFRP1 for predicting the risk of exacerbation was 0.847 (95% CI: 0.775 to 0.920), 86.20%, and 80.00%. The AUC, sensitivity, and specificity of CC16 for predicting the risk of exacerbation were 0.795 (95% CI: 0.711 to 0.879), 74.10%, and 86.20%. Conclusions These findings suggest that SFRP1 and CC16 may be useful serum markers for predicting the risk of exacerbation in COPD patients.
Collapse
|
9
|
Kayalar O, Oztay F, Yildirim M, Ersen E. Dysregulation of E-cadherin in pulmonary cell damage related with COPD contributes to emphysema. Toxicol Ind Health 2022; 38:330-341. [DOI: 10.1177/07482337221095638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Air pollution, especially at chronic exposure to high concentrations, is a respiratory risk factor for the development of chronic obstructive pulmonary disease (COPD). E-cadherin, a cell–cell adhesion protein, is involved in the integrity of the alveolar epithelium. Causes of E-cadherin decreases in emphysematous areas with pulmonary cell damage related to COPD are not well understood. We aimed to determine the molecules causing the decrease of E-cadherin and interactions between these molecules. In emphysematous and non-emphysematous areas of lungs from COPD patients (n = 35), levels of E-cadherin, HDACs, Snail, Zeb1, active-β-catenin, p120ctn, and Kaiso were determined by using Western Blot. The interactions of HDAC1, HDAC2, and p120ctn with transcription co-activators and Kaiso were examined by co-immunoprecipitation experiments. The methylation status of the CDH1 promoter was investigated. E-cadherin, Zeb1, Kaiso, and active-β-catenin were decreased in emphysema, while HDAC1, HDAC2, and p120ctn2 were increased. Snail, Zeb1, Twist, active-β-catenin, Kaiso, and p120ctn co-precipitated with HDAC1 and HDAC2. E-cadherin, Kaiso, and active-β-catenin co-precipitated with p120ctn. HDAC1–Snail and HDAC2–Kaiso interactions were increased in emphysema, but p120ctn-E-cadherin interaction was decreased. The results show that HDAC1–Snail and HDAC2–Kaiso interactions are capable of decreasing the E-cadherin in emphysema. The decreased interaction of p120ctn/E-cadherin leads to E-cadherin destruction. The decreased E-cadherin and its induced degradation in pneumocytes cause impaired repair and disintegrity of the epithelium. Approaches to suppress HDAC1–Snail and HDAC2–Kaiso interactions may help the protection of alveolar epithelial integrity by increasing the E-cadherin stability in pneumocytes.
Collapse
Affiliation(s)
- Ozgecan Kayalar
- Science Faculty, Department of Biology, Istanbul University, Istanbul, Turkey
- School of Medicine, Koc University Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul, Turkey
| | - Fusun Oztay
- Science Faculty, Department of Biology, Istanbul University, Istanbul, Turkey
| | - Merve Yildirim
- Science Faculty, Department of Biology, Istanbul University, Istanbul, Turkey
| | - Ezel Ersen
- Department of Chest Surgery, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
10
|
Rai N, Arteaga-Solis E, Goldklang M, Zelonina T, D’Armiento J. The Role of Secreted Frizzled-related Protein-1 in Allergic Asthma. Am J Respir Cell Mol Biol 2022; 66:293-301. [PMID: 34929134 PMCID: PMC8937247 DOI: 10.1165/rcmb.2020-0314oc] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/17/2021] [Indexed: 11/24/2022] Open
Abstract
Although allergic asthma is a highly prevalent chronic inflammatory condition, the underlying pathogenesis driving T-helper cell type 2 inflammation is not well understood. Wnt/β-catenin signaling has been implicated, but the influence of individual members of the pathway is not clear. We hypothesized that SFRP-1 (secreted frizzled-related protein-1), a Wnt signaling modulator, plays an important role in the development of allergic inflammation in asthma. Using an in vivo house dust mite asthma model, SFRP-1-/- mice were sensitized, and their BAL fluid was collected to evaluate airway inflammation. SFRP-1-/- mice exhibited less inflammation with reduced cellular infiltration and concentration of IL-5 in bronchoalveolar lavage fluid compared with wild-type (WT) mice. Similar findings were observed in WT mice treated with SFRP-1 inhibitor, WAY316606. Alveolar macrophages from sensitized SFRP-1-/- mice demonstrated reduced alternative polarization compared with WT, indicating that macrophages could mediate the alteration in inflammation seen in these mice. These findings suggest that SFRP-1 is an important potentiator of asthmatic airway inflammation.
Collapse
Affiliation(s)
| | | | | | | | - Jeanine D’Armiento
- Department of Anesthesiology
- Department of Medicine, and
- Department of Physiology and Cellular Biophysics, Columbia University, New York, New York
| |
Collapse
|
11
|
Secreted Frizzled-Related Protein 1 Promotes Odontoblastic Differentiation and Reparative Dentin Formation in Dental Pulp Cells. Cells 2021; 10:cells10092491. [PMID: 34572140 PMCID: PMC8468928 DOI: 10.3390/cells10092491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 01/09/2023] Open
Abstract
Direct pulp capping is an effective treatment for preserving dental pulp against carious or traumatic pulp exposure via the formation of protective reparative dentin by odontoblast-like cells. Reparative dentin formation can be stimulated by several signaling molecules; therefore, we investigated the effects of secreted frizzled-related protein (SFRP) 1 that was reported to be strongly expressed in odontoblasts of newborn molar tooth germs on odontoblastic differentiation and reparative dentin formation. In developing rat incisors, cells in the dental pulp, cervical loop, and inner enamel epithelium, as well as ameloblasts and preodontoblasts, weakly expressed Sfrp1; however, Sfrp1 was strongly expressed in mature odontoblasts. Human dental pulp cells (hDPCs) showed stronger expression of SFRP1 compared with periodontal ligament cells and gingival cells. SFRP1 knockdown in hDPCs abolished calcium chloride-induced mineralized nodule formation and odontoblast-related gene expression and decreased BMP-2 gene expression. Conversely, SFRP1 stimulation enhanced nodule formation and expression of BMP-2. Direct pulp capping treatment with SFRP1 induced the formation of a considerable amount of reparative dentin that has a structure similar to primary dentin. Our results indicate that SFRP1 is crucial for dentinogenesis and is important in promoting reparative dentin formation in response to injury.
Collapse
|
12
|
Guan H, Zhang J, Luan J, Xu H, Huang Z, Yu Q, Gou X, Xu L. Secreted Frizzled Related Proteins in Cardiovascular and Metabolic Diseases. Front Endocrinol (Lausanne) 2021; 12:712217. [PMID: 34489867 PMCID: PMC8417734 DOI: 10.3389/fendo.2021.712217] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/23/2021] [Indexed: 11/13/2022] Open
Abstract
Abnormal gene expression and secreted protein levels are accompanied by extensive pathological changes. Secreted frizzled related protein (SFRP) family members are antagonistic inhibitors of the Wnt signaling pathway, and they were recently found to be involved in the pathogenesis of a variety of metabolic diseases, which has led to extensive interest in SFRPs. Previous reports highlighted the importance of SFRPs in lipid metabolism, obesity, type 2 diabetes mellitus and cardiovascular diseases. In this review, we provide a detailed introduction of SFRPs, including their structural characteristics, receptors, inhibitors, signaling pathways and metabolic disease impacts. In addition to summarizing the pathologies and potential molecular mechanisms associated with SFRPs, this review further suggests the potential future use of SFRPs as disease biomarkers therapeutic targets.
Collapse
Affiliation(s)
- Hua Guan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi’an, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Jin Zhang
- Department of Preventive Medicine, School of Stomatology, Fourth Military Medical University, Xi’an, China
| | - Jing Luan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi’an, China
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Hao Xu
- Institution of Basic Medical Science, Xi’an Medical University, Xi’an, China
| | - Zhenghao Huang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Xingchun Gou
- Shaanxi Key Laboratory of Brain Disorders & Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
- *Correspondence: Lixian Xu, ; Xingchun Gou,
| | - Lixian Xu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anethesiology, School of Stomatology, Fourth Military Medical University, Xi’an, China
- *Correspondence: Lixian Xu, ; Xingchun Gou,
| |
Collapse
|
13
|
Mehta M, Dhanjal DS, Paudel KR, Singh B, Gupta G, Rajeshkumar S, Thangavelu L, Tambuwala MM, Bakshi HA, Chellappan DK, Pandey P, Dureja H, Charbe NB, Singh SK, Shukla SD, Nammi S, Aljabali AA, Wich PR, Hansbro PM, Satija S, Dua K. Cellular signalling pathways mediating the pathogenesis of chronic inflammatory respiratory diseases: an update. Inflammopharmacology 2020; 28:795-817. [PMID: 32189104 DOI: 10.1007/s10787-020-00698-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Respiratory disorders, especially non-communicable, chronic inflammatory diseases, are amongst the leading causes of mortality and morbidity worldwide. Respiratory diseases involve multiple pulmonary components, including airways and lungs that lead to their abnormal physiological functioning. Several signaling pathways have been reported to play an important role in the pathophysiology of respiratory diseases. These pathways, in addition, become the compounding factors contributing to the clinical outcomes in respiratory diseases. A range of signaling components such as Notch, Hedgehog, Wingless/Wnt, bone morphogenetic proteins, epidermal growth factor and fibroblast growth factor is primarily employed by these pathways in the eventual cascade of events. The different aberrations in such cell-signaling processes trigger the onset of respiratory diseases making the conventional therapeutic modalities ineffective. These challenges have prompted us to explore novel and effective approaches for the prevention and/or treatment of respiratory diseases. In this review, we have attempted to deliberate on the current literature describing the role of major cell signaling pathways in the pathogenesis of pulmonary diseases and discuss promising advances in the field of therapeutics that could lead to novel clinical therapies capable of preventing or reversing pulmonary vascular pathology in such patients.
Collapse
Affiliation(s)
- Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia
| | - Daljeet S Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Delhi, G.T. Road (NH-1), Phagwara, 144411, Punjab, India
| | - Keshav R Paudel
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia
| | - Bhupender Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar-Delhi, G.T. Road (NH-1), Phagwara, 144411, Punjab, India
| | - Gaurav Gupta
- School of Phamacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | - S Rajeshkumar
- Saveetha Dental College and Hospitals, Saveetha University, SIMATS, Chennai, Tamilnadu, India
| | - Lakshmi Thangavelu
- Saveetha Dental College and Hospitals, Saveetha University, SIMATS, Chennai, Tamilnadu, India
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Coleraine, County Londonderry, BT52 1SA, Northern Ireland, UK
| | - Hamid A Bakshi
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Parijat Pandey
- Shri Baba Mastnath Institute of Pharmaceutical Sciences and Research, Baba Mastnath University, Rohtak, 124001, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, 12401, India
| | - Nitin B Charbe
- Departamento de Química Orgánica, Facultad de Química Y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña McKenna 4860, 7820436, Santiago, Macul, Chile
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara, Punjab, 144411, India
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Alaa A Aljabali
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Peter R Wich
- Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Philip M Hansbro
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road (NH-1), Phagwara, Punjab, 144411, India.
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
- Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) and School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW, 2308, Australia.
- School of Pharmaceutical Sciences, Shoolini University, Himachal Pradesh, Bajhol, Sultanpur, Solan, 173 229, India.
| |
Collapse
|
14
|
Cruz-Hernández CD, Cruz-Burgos M, Cortés-Ramírez SA, Losada-García A, Camacho-Arroyo I, García-López P, Langley E, González-Covarrubias V, Llaguno-Munive M, Albino-Sánchez ME, Cruz-Colín JL, Pérez-Plasencia C, Beltrán-Anaya FO, Rodríguez-Dorantes M. SFRP1 increases TMPRSS2-ERG expression promoting neoplastic features in prostate cancer in vitro and in vivo. Cancer Cell Int 2020; 20:312. [PMID: 32694934 PMCID: PMC7364616 DOI: 10.1186/s12935-020-01333-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022] Open
Abstract
Background Prostate cancer (PCa) is the second cause of cancer related death in North American men. Androgens play an important role in its progression by regulating the expression of several genes including fusion ones that results from structural chromosome rearrangements. TMPRSS2-ERG is a fusion gene commonly observed in over 50% of PCa tumors, and its expression can be transcriptionally regulated by the androgen receptor (AR) given its androgen responsive elements. TMPRSS2-ERG could be involved in epithelial–mesenchymal transition (EMT) during tumor development. ERG has been reported as a key transcriptional factor in the AR-ERG-WNT network where five SFRP proteins, structurally similar to WNT ligands and considered to be WNT pathway antagonists, can regulate signaling in the extracellular space by binding to WNT proteins or Frizzled receptors. It has been shown that over-expression of SFRP1 protein can regulate the transcriptional activity of AR and inhibits the formation of colonies in LNCaP cells. However, the effect of SFRP1 has been controversial since differential effects have been observed depending on its concentration and tissue location. In this study, we explored the role of exogenous SFRP1 protein in cells expressing the TMPRSS2-ERG fusion. Methods To evaluate the effect of exogenous SFRP1 protein on PCa cells expressing TMPRSS2-ERG, we performed in silico analysis from TCGA cohort, expression assays by RT-qPCR and Western blot, cell viability and cell cycle measurements by cytometry, migration and invasion assays by xCELLigance system and murine xenografts. Results We demonstrated that SFRP1 protein increased ERG expression by promoting cellular migration in vitro and increasing tumor growth in vivo in PCa cells with the TMPRSS2-ERG fusion. Conclusions These results suggest the possible role of exogenous SFRP1 protein as a modulator of AR-ERG-WNT signaling network in cells positive to TMPRSS2-ERG. Further, investigation is needed to determine if SFRP1 protein could be a target in against this type of PCa.
Collapse
Affiliation(s)
- Carlos D Cruz-Hernández
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | - Marian Cruz-Burgos
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | - Sergio A Cortés-Ramírez
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | - Alberto Losada-García
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México; (UNAM), 04510 Mexico City, Mexico
| | | | | | | | | | - Martha E Albino-Sánchez
- Departamento de Biología celular, CINVESTAV, Av Instituto Politécnico Nacional 2508, San Pedro Zacatenco, 07360 Mexico city, Mexico
| | - José L Cruz-Colín
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | | | - Fredy O Beltrán-Anaya
- Instituto Nacional de Medicina Genómica, Périferico Sur 4809, Arenal Tepepan, 14610 Mexico city, Mexico
| | | |
Collapse
|
15
|
Noël A, Hansen S, Zaman A, Perveen Z, Pinkston R, Hossain E, Xiao R, Penn A. In utero exposures to electronic-cigarette aerosols impair the Wnt signaling during mouse lung development. Am J Physiol Lung Cell Mol Physiol 2020; 318:L705-L722. [PMID: 32083945 DOI: 10.1152/ajplung.00408.2019] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Currently, more than 9 million American adults, including women of childbearing age, use electronic-cigarettes (e-cigs). Further, the prevalence of maternal vaping now approaching 10% is similar to that of maternal smoking. Little, however, is known about the effects of fetal exposures to nicotine-rich e-cig aerosols on lung development. In this study, we assessed whether in utero exposures to e-cig aerosols compromised lung development in mice. A third-generation e-cig device was used to expose pregnant BALB/c mice by inhalation to 36 mg/mL of nicotine cinnamon-flavored e-cig aerosols for 14-31 days. This included exposures for either 12 days before mating plus during gestation (preconception groups) or only during gestation (prenatal groups). Respective control mice were exposed to filtered air. Subgroups of offspring were euthanized at birth or at 4 wk of age. Compared with respective air-exposed controls, both preconception and prenatal exposures to e-cig aerosols significantly decreased the offspring birth weight and body length. In the preconception group, 7 inflammation-related genes were downregulated, including 4 genes common to both dams and fetuses, denoting an e-cig immunosuppressive effect. Lung morphometry assessments of preconception e-cig-exposed offspring showed a significantly increased tissue fraction at birth. This result was supported by the downregulation of 75 lung genes involved in the Wnt signaling, which is essential to lung organogenesis. Thus, our data indicate that maternal vaping impairs pregnancy outcomes, alters fetal lung structure, and dysregulates the Wnt signaling. This study provides experimental evidence for future regulations of e-cig products for pregnant women and developmentally vulnerable populations.
Collapse
Affiliation(s)
- Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Shannon Hansen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Anusha Zaman
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Zakia Perveen
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rakeysha Pinkston
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana.,Health Research Center, Department of Environmental Toxicology, Southern University and A&M College, Baton Rouge, Louisiana
| | - Ekhtear Hossain
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| | - Rui Xiao
- Department of Anesthesiology, Columbia University Medical Center, New York, New York
| | - Arthur Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
16
|
Zhou M, Jiao L, Liu Y. sFRP2 promotes airway inflammation and Th17/Treg imbalance in COPD via Wnt/β-catenin pathway. Respir Physiol Neurobiol 2019; 270:103282. [PMID: 31430541 DOI: 10.1016/j.resp.2019.103282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 08/16/2019] [Accepted: 08/16/2019] [Indexed: 01/09/2023]
Abstract
Imbalance between inflammatory Th17 cells and immunosuppressive regulatory T cells (Treg) contributes to the progression of chronic obstructive pulmonary disease (COPD). We aims to investigate roles and mechanisms of secreted frizzled-related protein 2 (sFRP2) in airway inflammation and Th17/Treg differentiation in COPD. sFRP2 was significantly upregulated in the serum of patients with COPD and in human bronchial epithelial (HBE) cells that were exposed to cigarette smoke extract (CSE). sFRP2 was negatively correlated with FEV1/FVC. CSE increased IL-6 and TNF-α in HBE cells, which was reversed by sFRP2 silencing. CSE exposure elevated the percentage of Th17 in CD3+ CD8- cells while reduced the percentage of Treg in CD4+CD25+ cells. Knockdown of sFRP2 in peripheral blood mononuclear cells (PBMCs) attenuated Th17 differentiation and induced Treg differentiation. CSE suppressed the expression of β-catenin and Cyclin D1 in PBMCs while knockdown of sFRP2 markedly reversed the inhibitory effects of CSE. Wnt/β-catenin inhibition by Dickkopf-1 reversed the inhibitory effect of si-sFRP2 on the production of inflammatory cytokines and imbalance between Th17 and Treg cells caused by CSE. CSE induced sFRP2 potentiated airway inflammation and disturbed Th17/Treg homeostasis by inhibiting β-catenin.
Collapse
Affiliation(s)
- Miao Zhou
- Department of lung disease division, Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450008, China.
| | - Li Jiao
- Department of lung disease division, Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, 450008, China
| | - Yuanyuan Liu
- College of Acupuncture-moxibustion and Tuina, Henan University of Chinese Medicine, Zhengzhou, 450008, China
| |
Collapse
|
17
|
Cosin-Roger J, Ortiz-Masià MD, Barrachina MD. Macrophages as an Emerging Source of Wnt Ligands: Relevance in Mucosal Integrity. Front Immunol 2019; 10:2297. [PMID: 31608072 PMCID: PMC6769121 DOI: 10.3389/fimmu.2019.02297] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
The Wnt signaling pathway is a conserved pathway involved in important cellular processes such as the control of embryonic development, cellular polarity, cellular migration, and cell proliferation. In addition to playing a central role during embryogenesis, this pathway is also an essential part of adult homeostasis. Indeed, it controls the proliferation of epithelial cells in different organs such as intestine, lung, and kidney, and guarantees the maintenance of the mucosa in physiological conditions. The origin of this molecular pathway is the binding between Wnt ligands (belonging to a family of 19 different homologous secreted glycoproteins) and their specific membrane receptors, from the Frizzled receptor family. This specific interaction triggers the activation of the signaling cascade, which in turn activates or suppresses the expression of different genes in order to change the behavior of the cell. On the other hand, alterations of this pathway have been described in pathological conditions such as inflammation, fibrosis, and cancer. In recent years, macrophages-among other cell types-have emerged as a potential source of Wnt ligands. Due to their high plasticity, macrophages, which are central to the innate immune response, are capable of adopting different phenotypes depending on their microenvironment. In the past, two different phenotypes were described: a proinflammatory phenotype-M1 macrophages-and an anti-inflammatory phenotype-M2 macrophages-and a selective expression of Wnt ligands has been associated with said phenotypes. However, nowadays it is assumed that macrophages in vivo move through a continual spectrum of functional phenotypes. In both physiological and pathological (inflammation, fibrosis and cancer) conditions, the accumulation and polarization of macrophages conditions the future of the tissue, facilitating various scenarios, such as resolution of inflammation, activation of fibrosis, and cancer development due to the modulation of the Wnt signaling pathway, in autocrine and paracrine manner. In this work, we provide an overview of studies that have explored the role of macrophages and how they act as a source of Wnt ligands and as mediators of mucosal integrity.
Collapse
Affiliation(s)
| | - Mª Dolores Ortiz-Masià
- Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Mª Dolores Barrachina
- Departamento de Farmacología and CIBER, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| |
Collapse
|
18
|
Qu J, Yue L, Gao J, Yao H. Perspectives on Wnt Signal Pathway in the Pathogenesis and Therapeutics of Chronic Obstructive Pulmonary Disease. J Pharmacol Exp Ther 2019; 369:473-480. [PMID: 30952680 PMCID: PMC6538889 DOI: 10.1124/jpet.118.256222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 04/04/2019] [Indexed: 12/16/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic lung disease with progressive airflow limitation and functional decline. The pathogenic mechanisms for this disease include oxidative stress, inflammatory responses, disturbed protease/antiprotease equilibrium, apoptosis/proliferation imbalance, senescence, autophagy, metabolic reprogramming, and mitochondrial dysfunction. The Wnt signaling pathway is an evolutionarily conserved signaling pathway that is abnormal in COPD, including chronic bronchitis and pulmonary emphysema. Furthermore, Wnt signaling has been shown to modulate aforementioned cellular processes involved in COPD. From this perspective, we provide an updated understanding of the crosstalk between Wnt signal and these cellular processes, and highlight the crucial role of the Wnt signal during the development of COPD. We also discuss the potential for targeting the Wnt signal in future translational and pharmacological therapeutics aimed at prevention and treatment of this disease.
Collapse
Affiliation(s)
- Jiao Qu
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Li Yue
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Jian Gao
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| | - Hongwei Yao
- The Second Affiliated Hospital, School of Pharmacy, Dalian Medical University, Dalian, Liaoning, China (J. Q., J. G.); The First Affiliated Hospital, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China (J.Q., J.G.); Department of Orthopedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island (L.Y.); and Department of Molecular Biology, Cell Biology and Biochemistry, Brown University Division of Biology and Medicine, Providence, Rhode Island (H.Y.)
| |
Collapse
|
19
|
Zhou J, Yi Z, Fu Q. Dynamic decreased expression and hypermethylation of secreted frizzled-related protein 1 and 4 over the course of pulmonary fibrosis in mice. Life Sci 2019; 218:241-252. [PMID: 30586565 DOI: 10.1016/j.lfs.2018.12.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/18/2018] [Accepted: 12/22/2018] [Indexed: 01/08/2023]
Abstract
Aberrantly activated Wnt signaling pathway and dysregulation of extracellular antagonists of Wnt signaling have been revealed in pulmonary fibrosis. In this study we evaluated the expression of secreted frizzled-related proteins (SFRPs) and their aberrant promoter methylation to investigate the involvement of epigenetic regulation in pulmonary fibrosis. The pulmonary fibrosis induced by intratracheal injection of bleomycin (BLM) into mice was adopted. The transcription and relative protein expression of SFRPs were detected at Day 7 (D7), D14, and D21. DNA methylation analysis was performed by methylation-specific polymerase chain reaction (MSP). A DNA methyltransferase (DNMT) inhibitor (5-aza-2'-deoxycytidine; 5-aza) was used for demethylation and the relative β-catenin expression levels were measured to assess overactivity of the canonical Wnt signaling pathway. The transcription and protein expression of SFRP1 significantly decreased at D14 and D21, whereas the transcription and protein expression of SFRP4 significantly decreased at D7 and stayed downregulated until D21. The significantly hypermethylated promoters of SFRP1 and SFRP4 resulted in impaired transcription and decreased expression during pulmonary fibrosis in mice. Besides, reactivation of SFRP1 and SFRP4 by 5-aza reduced β-catenin mRNA and protein expression in vivo and in vitro. Animal experiments confirmed that 5-aza could significantly alleviate bleomycin-induced pulmonary fibrosis in mice. Thus, changes of promoter hypermethylation might downregulate SFRP1 and SFRP4 at different stages of pulmonary fibrosis, and the finding supports the usefulness of DNMT inhibitors, which might effectively reverse activation of β-catenin and reduce pulmonary fibrosis in mice. These data provide a possible new direction in the research on pulmonary fibrosis treatments.
Collapse
Affiliation(s)
- Junfei Zhou
- Department of Rheumatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Zheng Yi
- Department of Rheumatology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, PR China.
| | - Qiang Fu
- Department of Rheumatology, The First Affiliated Hospital of University of South China, HengYang 421001, PR China
| |
Collapse
|
20
|
Peng JX, Liang SY, Li L. sFRP1 exerts effects on gastric cancer cells through GSK3β/Rac1‑mediated restraint of TGFβ/Smad3 signaling. Oncol Rep 2018; 41:224-234. [PMID: 30542739 PMCID: PMC6278527 DOI: 10.3892/or.2018.6838] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
Secreted frizzled-related protein 1 (sFRP1) is an inhibitor of canonical Wnt signaling; however, previous studies have determined a tumor-promoting function of sFRP1 in a number of different cancer types. A previous study demonstrated that sFRP1 overexpression was associated with an aggressive phenotype and the activation of transforming growth factor β (TGFβ) signaling. sFRP1 overexpression and sFRP1 knockdown cell models were established. Immunoblotting was conducted to examine the protein levels of the associated molecules. Immunofluorescence staining followed by confocal microscopy was performed to visualize the cytoskeleton alterations and subcellular localization of key proteins. sFRP1 overexpression restored glycogen synthase kinase 3β (GSK3β) activity, which activated Rac family small GTPase 1 (Rac1). GSK3β and Rac1 mediated the effect of sFRP1 on the positive regulation of cell growth and migration/invasion. Inhibition of GSK3β or Rac1 abolished the regulation of sFRP1 on TGFβ/SMAD family member 3 (Smad3) signaling and the aggressive phenotype; however, GSK3β or Rac1 overexpression increased cell migration/invasion and restrained Smad3 activity by preventing its nuclear translocation and limiting its transcriptional activity. The present study demonstrated a tumor-promoting function of sFRP1-overexpression by selectively activating TGFβ signaling in gastric cancer cells. GSK3β and Rac1 serve an important function in mediating the sFRP1-induced malignant alterations and signaling changes.
Collapse
Affiliation(s)
- Ji-Xiang Peng
- Department of Gastrointestinal Surgery, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| | - Shun-Yu Liang
- Department of Gastrointestinal Surgery, Guangzhou First Municipal People's Hospital, Affiliated Guangzhou Medical College, Guangzhou, Guangdong 510180, P.R. China
| | - Li Li
- Department of Gastrointestinal Surgery, Guangzhou First People's Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou, Guangdong 510180, P.R. China
| |
Collapse
|
21
|
Skronska-Wasek W, Gosens R, Königshoff M, Baarsma HA. WNT receptor signalling in lung physiology and pathology. Pharmacol Ther 2018; 187:150-166. [PMID: 29458107 DOI: 10.1016/j.pharmthera.2018.02.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The WNT signalling cascades have emerged as critical regulators of a wide variety of biological aspects involved in lung development as well as in physiological and pathophysiological processes in the adult lung. WNTs (secreted glycoproteins) interact with various transmembrane receptors and co-receptors to activate signalling pathways that regulate transcriptional as well as non-transcriptional responses within cells. In physiological conditions, the majority of WNT receptors and co-receptors can be detected in the adult lung. However, dysregulation of WNT signalling pathways contributes to the development and progression of chronic lung pathologies, including idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD), asthma and lung cancer. The interaction between a WNT and the (co-)receptor(s) present at the cell surface is the initial step in transducing an extracellular signal into an intracellular response. This proximal event in WNT signal transduction with (cell-specific) ligand-receptor interactions is of great interest as a potential target for pharmacological intervention. In this review we highlight the diverse expression of various WNT receptors and co-receptors in the aforementioned chronic lung diseases and discuss the currently available biologicals and pharmacological tools to modify proximal WNT signalling.
Collapse
Affiliation(s)
- Wioletta Skronska-Wasek
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany
| | - Reinoud Gosens
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Hoeke Abele Baarsma
- Comprehensive Pneumology Center, Research Unit Lung Repair and Regeneration, Helmholtz Center Munich, Member of the German Center for Lung Research, Ludwig Maximilians University Munich, University Hospital Grosshadern, Munich, Germany; GRIAC Research Institute, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
22
|
Abstract
Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF), represent a significant and increasing health burden. Current therapies are largely symptomatic, and novel therapeutic approaches are needed. Aging has emerged as a contributing factor for the development of both IPF and COPD because their prevalence increases with age, and several pathological features of these diseases resemble classical hallmarks of aging. Aging is thought to be driven in part by aberrant activity of developmental signaling pathways that thus might drive pathological changes, a process termed antagonistic pleiotropy or developmental drift. The developmental WNT pathway is fundamental for lung development, and altered WNT activity has been reported to contribute to the pathogenesis of CLD, in particular to COPD and IPF. Although to date only limited data on WNT signaling during lung aging exist, WNT signal regulation during aging and its effects on age-related pathologies in other organs have recently been investigated. In this review, we discuss evidence of dysregulated WNT signaling in CLD in the context of WNT signal alteration in organ aging and its potential impact on age-related cellular mechanisms, such as senescence or stem cell exhaustion.
Collapse
|
23
|
Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochim Biophys Acta Mol Basis Dis 2017; 1863:3226-3242. [PMID: 28866134 DOI: 10.1016/j.bbadis.2017.08.031] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/10/2017] [Accepted: 08/29/2017] [Indexed: 12/23/2022]
Abstract
Embryonic lung development requires reciprocal endodermal-mesodermal interactions; mediated by various signaling proteins. Wnt/β-catenin is a signaling protein that exhibits the pivotal role in lung development, injury and repair while aberrant expression of Wnt/β-catenin signaling leads to asthmatic airway remodeling: characterized by hyperplasia and hypertrophy of airway smooth muscle cells, alveolar and vascular damage goblet cells metaplasia, and deposition of extracellular matrix; resulting in decreased lung compliance and increased airway resistance. The substantial evidence suggests that Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Here, we summarized the recent advances related to the mechanistic role of Wnt/β-catenin signaling in lung development, consequences of aberrant expression or deletion of Wnt/β-catenin signaling in expansion and progression of asthmatic airway remodeling, and linking early-impaired pulmonary development and airway remodeling later in life. Finally, we emphasized all possible recent potential therapeutic significance and future prospectives, that are adaptable for therapeutic intervention to treat asthmatic airway remodeling.
Collapse
Affiliation(s)
- Musaddique Hussain
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| | - Chengyun Xu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China
| | - Meiping Lu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Xiling Wu
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China.
| | - Lanfang Tang
- Department of Respiratory Medicine, the Affiliated Children Hospital, School of Medicine, Zhejiang University, Hangzhou City 310006, China
| | - Ximei Wu
- Department of Pharmacology, School of Medicine, Zhejiang University, Hangzhou City 310058, China; The Key Respiratory Drug Research Laboratory of China Food and Drug Administration, School of Medicine, Zhejiang University, Hangzhou City 310058, China.
| |
Collapse
|
24
|
Sharma A, Yang WL, Ochani M, Wang P. Mitigation of sepsis-induced inflammatory responses and organ injury through targeting Wnt/β-catenin signaling. Sci Rep 2017; 7:9235. [PMID: 28835626 PMCID: PMC5569053 DOI: 10.1038/s41598-017-08711-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/13/2017] [Indexed: 12/29/2022] Open
Abstract
The Wnt/β-catenin pathway has been involved in regulating inflammation in various infectious and inflammatory diseases. Sepsis is a life-threatening condition caused by dysregulated inflammatory response to infection with no effective therapy available. Recently elevated Wnt/β-catenin signaling has been detected in sepsis. However, its contribution to sepsis-associated inflammatory response remains to be explored. In this study, we show that inhibition of Wnt/β-catenin signaling reduces inflammation and mitigates sepsis-induced organ injury. Using in vitro LPS-stimulated RAW264.7 macrophages, we demonstrate that a small-molecule inhibitor of β-catenin responsive transcription, iCRT3, significantly reduces the LPS-induced Wnt/β-catenin activity and also inhibits TNF-α production and IκB degradation in a dose-dependent manner. Intraperitoneal administration of iCRT3 to C57BL/6 mice, subjected to cecal ligation and puncture-induced sepsis, decreases the plasma levels of proinflammatory cytokines and organ injury markers in a dose-dependent manner. The histological integrity of the lungs is improved with iCRT3 treatment, along with reduced lung collagen deposition and apoptosis. In addition, iCRT3 treatment also decreases the expression of the cytokines, neutrophil chemoattractants, as well as the MPO activity in the lungs of septic mice. Based on these findings we conclude that targeting the Wnt/β-Catenin pathway may provide a potential therapeutic approach for treatment of sepsis.
Collapse
Affiliation(s)
- Archna Sharma
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Weng-Lang Yang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
- Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, NY, 11030, USA
| | - Mahendar Ochani
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, NY, 11030, USA.
- Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, NY, 11030, USA.
| |
Collapse
|
25
|
Distinct Roles of Wnt/ β-Catenin Signaling in the Pathogenesis of Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Mediators Inflamm 2017; 2017:3520581. [PMID: 28588349 PMCID: PMC5447271 DOI: 10.1155/2017/3520581] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/29/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
Wnt signaling pathways are tightly controlled under a physiological condition, under which they play key roles in many biological functions, including cell fate specification and tissue regeneration. Increasing lines of evidence recently demonstrated that a dysregulated activation of Wnt signaling, particularly the Wnt/β-catenin signaling, was involved in the pathogenesis of chronic pulmonary diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). In this respect, Wnt signaling interacts with other cellular signaling pathways to regulate the initiation and pathogenic procedures of airway inflammation and remodeling, pulmonary myofibroblast proliferation, epithelial-to-mesenchymal transition (EMT), and development of emphysema. Intriguingly, Wnt/β-catenin signaling is activated in IPF; an inhibition of this signaling leads to an alleviation of pulmonary inflammation and fibrosis in experimental models. Conversely, Wnt/β-catenin signaling is inactivated in COPD tissues, and its reactivation results in an amelioration of airspace enlargement with a restored alveolar epithelial structure and function in emphysema models. These studies thus imply distinct mechanisms of Wnt/β-catenin signaling in the pathogenesis of these two chronic pulmonary diseases, indicating potential targets for COPD and IPF treatments. This review article aims to summarize the involvement and pathogenic roles of Wnt signaling pathways in the COPD and IPF, with a focus on the implication of Wnt/β-catenin signaling as underlying mechanisms and therapeutic targets in these two incurable diseases.
Collapse
|
26
|
Unachukwu U, Trischler J, Goldklang M, Xiao R, D'Armiento J. Maternal smoke exposure decreases mesenchymal proliferation and modulates Rho-GTPase-dependent actin cytoskeletal signaling in fetal lungs. FASEB J 2017; 31:2340-2351. [PMID: 28209772 DOI: 10.1096/fj.201601063r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/30/2017] [Indexed: 12/12/2022]
Abstract
The present study tested the hypothesis that maternal smoke exposure results in fetal lung growth retardation due to dysregulation in various signaling pathways, including the Wnt (wingless-related integration site)/β-catenin pathway. Pregnant female C57BL/6J mice were exposed to cigarette smoke (100-150 mg/m3) or room air, and offspring were humanely killed on 12.5, 14.5, 16.5, and 18.5 d post coitum (dpc). We assessed lung stereology with Cavalieri estimation; apoptosis with proliferating cell nuclear antigen, TUNEL, and caspase assays; and gene expression with quantitative PCR (qPCR) and RNA sequencing on lung epithelium and mesenchyme retrieved by laser capture microdissection. Results demonstrated a significant decrease in body weight and lung volume of smoke-exposed embryos. At 16.5 dpc, the reduction in lung volume was due to loss of lung mesenchymal tissue correlating with a decrease in cell proliferation (n = 10; air: 61.65% vs. smoke: 44.21%, P < 0.05). RNA sequence analysis demonstrated an alteration in the Wnt pathway, and qPCR confirmed an increased expression of secreted frizzled-related protein 1 (sFRP-1) [n = 12; relative quantification (RQ) 1 vs. 2.33, P < 0.05] and down-regulation of Cyclin D1 (n = 7; RQ 1 vs. 0.61, P < 0.05) in mesenchymal tissue. Furthermore, genome expression studies revealed a smoke-induced up-regulation of Rho-GTPase-dependent actin cytoskeletal signaling that can lead to loss of tissue integrity.-Unachukwu, U., Trischler, J., Goldklang, M., Xiao, R., D'Armiento, J. Maternal smoke exposure decreases mesenchymal proliferation and modulates Rho-GTPase-dependent actin cytoskeletal signaling in fetal lungs.
Collapse
Affiliation(s)
- Uchenna Unachukwu
- Center for Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jordis Trischler
- Center for Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Monica Goldklang
- Center for Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Rui Xiao
- Center for Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Jeanine D'Armiento
- Center for Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| |
Collapse
|
27
|
Kim BA, Jee HG, Yi JW, Kim SJ, Chai YJ, Choi JY, Lee KE. Expression Profiling of a Human Thyroid Cell Line Stably Expressing the BRAFV600E Mutation. Cancer Genomics Proteomics 2017; 14:53-67. [PMID: 28031237 DOI: 10.21873/cgp.20018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/05/2016] [Accepted: 12/06/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND/AIM The BRAFV600E mutation acts as an initiator of cancer development in papillary thyroid carcinoma (PTC). Gene expression changes caused by the BRAFV600E mutation may have an important role in thyroid cancer development. MATERIALS AND METHODS To study genomic alterations caused by the BRAFV600E mutation, we made human thyroid cell lines that harbor the wild-type BRAF gene (Nthy/WT) and the V600E mutant-type BRAF gene (Nthy/V600E). RESULTS Flow cytometry and western blotting showed stable transfection of the BRAF gene. In functional experiments, Nthy/V600E showed increased anchorage-independent growth and invasion through Matrigel, compared to Nthy/WT. Microarray analysis revealed that 2,441 genes were up-regulated in Nthy/V600E compared to Nthy/WT. Gene ontology analysis showed that the up-regulated genes were associated with cell adhesion, migration, and the ERK and MAPK cascade, and pathway analysis showed enrichment in cancer-related pathways. CONCLUSION Our Nthy/WT and Nthy/V600E cell line pair could be a suitable model to study the molecular characteristics of BRAFV600E PTC.
Collapse
Affiliation(s)
- Byoung-Ae Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyeon-Gun Jee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jin Wook Yi
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Su-Jin Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Young Jun Chai
- Department of Surgery, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - June Young Choi
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Kyu Eun Lee
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea .,Department of Surgery, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
Ota C, Baarsma HA, Wagner DE, Hilgendorff A, Königshoff M. Linking bronchopulmonary dysplasia to adult chronic lung diseases: role of WNT signaling. Mol Cell Pediatr 2016; 3:34. [PMID: 27718180 PMCID: PMC5055515 DOI: 10.1186/s40348-016-0062-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Bronchopulmonary dysplasia (BPD) is one of the most common chronic lung diseases in infants caused by pre- and/or postnatal lung injury. BPD is characterized by arrested alveolarization and vascularization due to extracellular matrix remodeling, inflammation, and impaired growth factor signaling. WNT signaling is a critical pathway for normal lung development, and its altered signaling has been shown to be involved in the onset and progression of incurable chronic lung diseases in adulthood, such as chronic obstructive pulmonary disease (COPD) or idiopathic pulmonary fibrosis (IPF). In this review, we summarize the impact of WNT signaling on different stages of lung development and its potential contribution to developmental lung diseases, especially BPD, and chronic lung diseases in adulthood.
Collapse
Affiliation(s)
- Chiharu Ota
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.
| | - Hoeke A Baarsma
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Darcy E Wagner
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| | - Anne Hilgendorff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany.,The Perinatal Center, Campus Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Melanie Königshoff
- Comprehensive Pneumology Center, Helmholtz Center Munich, Ludwig-Maximilians-University, University Hospital Grosshadern, German Center of Lung Research (DZL), Munich, Germany
| |
Collapse
|
29
|
Désert R, Mebarki S, Desille M, Sicard M, Lavergne E, Renaud S, Bergeat D, Sulpice L, Perret C, Turlin B, Clément B, Musso O. "Fibrous nests" in human hepatocellular carcinoma express a Wnt-induced gene signature associated with poor clinical outcome. Int J Biochem Cell Biol 2016; 81:195-207. [PMID: 27545991 DOI: 10.1016/j.biocel.2016.08.017] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 08/01/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the 3rd cause of cancer-related death worldwide. Most cases arise in a background of chronic inflammation, extracellular matrix (ECM) remodeling, severe fibrosis and stem/progenitor cell amplification. Although HCCs are soft cellular tumors, they may contain fibrous nests within the tumor mass. Thus, the aim of this study was to explore cancer cell phenotypes in fibrous nests. Combined anatomic pathology, tissue microarray and real-time PCR analyses revealed that HCCs (n=82) containing fibrous nests were poorly differentiated, expressed Wnt pathway components and target genes, as well as markers of stem/progenitor cells, such as CD44, LGR5 and SOX9. Consistently, in severe liver fibroses (n=66) and in HCCs containing fibrous nests, weighted correlation analysis revealed a gene network including the myofibroblast marker ACTA2, the basement membrane components COL4A1 and LAMC1, the Wnt pathway members FZD1; FZD7; WNT2; LEF1; DKK1 and the Secreted Frizzled Related Proteins (SFRPs) 1; 2 and 5. Moreover, unbiased random survival forest analysis of a transcriptomic dataset of 247 HCC patients revealed high DKK1, COL4A1, SFRP1 and LAMC1 to be associated with advanced tumor staging as well as with bad overall and disease-free survival. In vitro, these genes were upregulated in liver cancer stem/progenitor cells upon Wnt-induced mesenchymal commitment and myofibroblast differentiation. In conclusion, fibrous nests express Wnt target genes, as well as markers of cancer stem cells and mesenchymal commitment. Fibrous nests embody the specific microenvironment of the cancer stem cell niche and can be detected by routine anatomic pathology analyses.
Collapse
Affiliation(s)
- Romain Désert
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Sihem Mebarki
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Mireille Desille
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France; CHU Rennes, Centre de Ressources Biologiques Santé BB-0033-00056, Rennes, France.
| | - Marie Sicard
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Elise Lavergne
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Stéphanie Renaud
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Damien Bergeat
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France; CHU de Rennes, Dept. of Gastrointestinal and Hepatobiliary Surgery, Rennes, France.
| | - Laurent Sulpice
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France; CHU de Rennes, Dept. of Gastrointestinal and Hepatobiliary Surgery, Rennes, France.
| | - Christine Perret
- Inserm, U1016, Institut Cochin, Paris, France; Cnrs, UMR8104, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France.
| | - Bruno Turlin
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France; CHU Rennes, Centre de Ressources Biologiques Santé BB-0033-00056, Rennes, France.
| | - Bruno Clément
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| | - Orlando Musso
- Inserm, UMR991, Liver Metabolisms and Cancer, Rennes, France; Université de Rennes 1, F-35043 Rennes, France.
| |
Collapse
|
30
|
Martinez L, Gomez C, Vazquez-Padron RI. Age-related changes in monocytes exacerbate neointimal hyperplasia after vascular injury. Oncotarget 2016; 6:17054-64. [PMID: 25965835 PMCID: PMC4627291 DOI: 10.18632/oncotarget.3881] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 03/31/2015] [Indexed: 01/09/2023] Open
Abstract
Neointimal hyperplasia is the leading cause of restenosis after endovascular interventions. It is characterized by the accumulation of myofibroblast-like cells and extracellular matrix in the innermost layer of the wall and is exacerbated by inflammation. Monocytes from either young or aged rats were applied perivascularly to injured vascular walls of young recipient animals. Monocytes from aged rats, but not young donors, increased neointima thickness. Accordingly, the gene expression profiles of CD11b+ monocytes from aged rats showed significant up-regulation of genes involved in cellular adhesion, lipid degradation, cytotoxicity, differentiation, and inflammation. These included cadherin 13 (Cdh13), colony stimulating factor 1 (Csf1), chemokine C-X-C motif ligand 1 (Cxcl1), endothelial cell-selective adhesion molecule (Esam), and interferon gamma (Ifng). In conclusion, our results suggest that the increased inflammatory and adhesive profile of monocytes contributes to pathological wall remodeling in aged-related vascular diseases.
Collapse
Affiliation(s)
- Laisel Martinez
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Camilo Gomez
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Roberto I Vazquez-Padron
- Department of Surgery and Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
31
|
Reuter S, Beckert H, Taube C. Take the Wnt out of the inflammatory sails: modulatory effects of Wnt in airway diseases. J Transl Med 2016; 96:177-85. [PMID: 26595171 DOI: 10.1038/labinvest.2015.143] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/08/2015] [Accepted: 09/28/2015] [Indexed: 12/11/2022] Open
Abstract
Bronchial asthma and chronic obstructive pulmonary disease (COPD) are chronic diseases that are associated with inflammation and structural changes in the airways and lungs. Recent findings have implicated Wnt pathways in critically regulating inflammatory responses, especially in asthma. Furthermore, canonical and noncanonical Wnt pathways are involved in structural changes such as airway remodeling, goblet cell metaplasia, and airway smooth muscle (ASM) proliferation. In COPD, Wnt pathways are not only associated with structural changes in the airways but also involved in the development of emphysema. The present review summarizes the role and function of the canonical and noncanonical Wnt pathway with regard to airway inflammation and structural changes in asthma and COPD. Further identification of the role and function of different Wnt molecules and pathways could help to develop novel therapeutic options for these diseases.
Collapse
Affiliation(s)
- Sebastian Reuter
- Priority Area Asthma and Allergy, Research Center Borstel, Airway Research Center North, Member of the German Center for Lung Research, Borstel, Germany
| | - Hendrik Beckert
- III Medical Clinic, University Medical Center, Mainz, Germany
| | - Christian Taube
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
32
|
Wang A, Zsengellér ZK, Hecht JL, Buccafusca R, Burke SD, Rajakumar A, Weingart E, Yu PB, Salahuddin S, Karumanchi SA. Excess placental secreted frizzled-related protein 1 in maternal smokers impairs fetal growth. J Clin Invest 2015; 125:4021-5. [PMID: 26413870 DOI: 10.1172/jci80457] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 08/17/2015] [Indexed: 12/18/2022] Open
Abstract
Maternal cigarette smoking during pregnancy remains one of the most common and preventable causes of fetal growth restriction (FGR), a condition in which a fetus is unable to achieve its genetically determined potential size. Even though epidemiologic evidence clearly links maternal cigarette smoking with FGR, insight into the molecular mechanisms of cigarette smoke-induced FGR is lacking. Here, we performed transcriptional profiling of placentas obtained from smoking mothers who delivered growth-restricted infants and identified secreted frizzled-related protein 1 (sFRP1), an extracellular antagonist of endogenous WNT signaling, as a candidate molecule. sFRP1 mRNA and protein levels were markedly upregulated (~10-fold) in placentas from smoking mothers compared with those from nonsmokers. In pregnant mice, adenovirus-mediated overexpression of sFRP1 led to FGR, increased karyorrhexis in the junctional zone, and decreased proliferation of labyrinthine trophoblasts. Consistent with our hypothesis that placental WNT signaling is suppressed in maternal smokers, we found that exposure to carbon monoxide analogs led to reduced WNT signaling, increased SFRP1 mRNA expression, and decreased cellular proliferation in a trophoblast cell line. Moreover, administration of carbon monoxide analogs to pregnant mice in late gestation led to FGR. In summary, our results indicate that the increased placental expression of sFRP1 seen in smokers impairs fetal growth by inhibiting WNT signaling and trophoblast proliferation.
Collapse
|
33
|
Platelet-derived Wnt antagonist Dickkopf-1 is implicated in ICAM-1/VCAM-1-mediated neutrophilic acute lung inflammation. Blood 2015; 126:2220-9. [PMID: 26351298 DOI: 10.1182/blood-2015-02-622233] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 08/20/2015] [Indexed: 12/14/2022] Open
Abstract
Neutrophil infiltration represents the early acute inflammatory response in acute lung injury. The recruitment of neutrophils from the peripheral blood across the endothelial-epithelial barrier into the alveolar airspace is highly regulated by the adhesion molecules on alveolar epithelial cells (AECs). Wnt/β-catenin signaling is involved in the progression of inflammatory lung diseases including asthma, emphysema, and pulmonary fibrosis. However, the function of Wnt/β-catenin signaling in acute lung inflammation is unknown. Here, we identified platelet-derived Dickkopf-1 (Dkk1) as the major Wnt antagonist contributing to the suppression of Wnt/β-catenin signaling in AECs during acute lung inflammation. Intratracheal administration of Wnt3a or an antibody capable of neutralizing Dkk1 inhibited neutrophil influx into the alveolar airspace of injured lungs. Activation of Wnt/β-catenin signaling in AECs attenuated intercellular adhesion molecule 1 (ICAM-1)/vascular cell adhesion molecule 1 (VCAM-1)-mediated adhesion of both macrophages and neutrophils to AECs. Our results suggest a role for Wnt/β-catenin signaling in modulating the inflammatory response, and a functional communication between platelets and AECs during acute lung inflammation. Targeting Wnt/β-catenin signaling and the communication between platelets and AECs therefore represents potential therapeutic strategies to limit the damage of acute pulmonary inflammation.
Collapse
|
34
|
Sklepkiewicz P, Shiomi T, Kaur R, Sun J, Kwon S, Mercer B, Bodine P, Schermuly RT, George I, Schulze PC, D'Armiento JM. Loss of secreted frizzled-related protein-1 leads to deterioration of cardiac function in mice and plays a role in human cardiomyopathy. Circ Heart Fail 2015; 8:362-72. [PMID: 25669938 DOI: 10.1161/circheartfailure.114.001274] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
BACKGROUND The Wnt/β-catenin signaling pathway plays a central role during cardiac development and has been implicated in cardiac remodeling and aging. However, the role of Wnt modulators in this process is unknown. In this study, we examined the role of the Wnt signaling inhibitor secreted frizzled-related protein-1 (sFRP-1) in aged wild-type and sFRP-1-deficient mice. METHODS AND RESULTS sFRP-1 gene deletion mice were grossly normal with no difference in mortality but developed abnormal cardiac structure and dysfunction with progressive age. Ventricular dilation and hypertrophy in addition to deterioration of cardiac function and massive cardiac fibrosis, all features present in dilated cardiomyopathy, were observed in the aged sFRP-1 knockout mice. Loss of sFRP-1 led to increased expression of Wnt ligands (Wnt1, 3, 7b, and 16) and Wnt target genes (Wisp1 and Lef1) in aged hearts, which correlated with increased protein levels of β-catenin. Cardiac fibroblasts lacking endogenous sFRP-1 showed increased α-smooth muscle actin expression, higher cell proliferation rates, and increased collagen production consistent with the cardiac phenotype exhibited in aged sFRP-1 knockout mice. The clinical relevance of these findings was supported by the demonstration of decreased sFRP-1 gene expression and increased Wisp-1 levels in the left ventricles of patients with ischemic dilated cardiomyopathy and dilated cardiomyopathy. CONCLUSIONS This study identifies a novel role of sFRP-1 in age-related cardiac deterioration and fibrosis. Further exploration of this pathway will identify downstream molecules important in these processes and also suggest the potential use of Wnt signaling agents as therapeutic targets for age-related cardiovascular disorders in humans.
Collapse
Affiliation(s)
- Piotr Sklepkiewicz
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Takayuki Shiomi
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Rajbir Kaur
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Jie Sun
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Susan Kwon
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Becky Mercer
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Peter Bodine
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Ralph Theo Schermuly
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Isaac George
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - P Christian Schulze
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.)
| | - Jeanine M D'Armiento
- From the Center for Molecular Pulmonary Disease, Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, NY (P.S., T.S., R.K., J.S., S.K., B.M., J.M.D.); Women's Health Research Institute, Department of Osteoporosis, Wyeth Research, Collegeville, PA (P.B.); Max Planck Institute, Department of Pathophysiology and Pulmonary, Bad Nauheim, Germany (R.T.S.); Center for Advanced Cardiac Care, Columbia University Medical Center, New York, NY (C.S.); and Division of Cardiothoracic Surgery, Department of Surgery, New York Presbyterian Hospital/Columbia University Medical Center, New York, NY (I.G.).
| |
Collapse
|
35
|
Moura RS, Carvalho-Correia E, daMota P, Correia-Pinto J. Canonical Wnt signaling activity in early stages of chick lung development. PLoS One 2014; 9:e112388. [PMID: 25460002 PMCID: PMC4251901 DOI: 10.1371/journal.pone.0112388] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Accepted: 10/15/2014] [Indexed: 01/08/2023] Open
Abstract
Wnt signaling pathway is an essential player during vertebrate embryonic development which has been associated with several developmental processes such as gastrulation, body axis formation and morphogenesis of numerous organs, namely the lung. Wnt proteins act through specific transmembrane receptors, which activate intracellular pathways that regulate cellular processes such as cell proliferation, differentiation and death. Morphogenesis of the fetal lung depends on epithelial-mesenchymal interactions that are governed by several growth and transcription factors that regulate cell proliferation, fate, migration and differentiation. This process is controlled by different signaling pathways such as FGF, Shh and Wnt among others. Wnt signaling is recognized as a key molecular player in mammalian pulmonary development but little is known about its function in avian lung development. The present work characterizes, for the first time, the expression pattern of several Wnt signaling members, such as wnt-1, wnt-2b, wnt-3a, wnt-5a, wnt-7b, wnt-8b, wnt-9a, lrp5, lrp6, sfrp1, dkk1, β-catenin and axin2 at early stages of chick lung development. In general, their expression is similar to their mammalian counterparts. By assessing protein expression levels of active/total β-catenin and phospho-LRP6/LRP6 it is revealed that canonical Wnt signaling is active in this embryonic tissue. In vitro inhibition studies were performed in order to evaluate the function of Wnt signaling pathway in lung branching. Lung explants treated with canonical Wnt signaling inhibitors (FH535 and PK115-584) presented an impairment of secondary branch formation after 48 h of culture along with a decrease in axin2 expression levels. Branching analysis confirmed this inhibition. Wnt-FGF crosstalk assessment revealed that this interaction is preserved in the chick lung. This study demonstrates that Wnt signaling is crucial for precise chick lung branching and further supports the avian lung as a good model for branching studies since it recapitulates early mammalian pulmonary development.
Collapse
Affiliation(s)
- Rute Silva Moura
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Biology Department, School of Sciences, University of Minho, Braga, Portugal
| | - Eduarda Carvalho-Correia
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paulo daMota
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Urology, Hospital de Braga, Braga, Portugal
| | - Jorge Correia-Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Pediatric Surgery, Hospital de Braga, Braga, Portugal
| |
Collapse
|
36
|
Slany A, Meshcheryakova A, Beer A, Ankersmit HJ, Paulitschke V, Gerner C. Plasticity of fibroblasts demonstrated by tissue-specific and function-related proteome profiling. Clin Proteomics 2014; 11:41. [PMID: 26029019 PMCID: PMC4448269 DOI: 10.1186/1559-0275-11-41] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 10/17/2014] [Indexed: 01/15/2023] Open
Abstract
Background Fibroblasts are mesenchymal stromal cells which occur in all tissue types. While their main function is related to ECM production and physical support, they are also important players in wound healing, and have further been recognized to be able to modulate inflammatory processes and support tumor growth. Fibroblasts can display distinct phenotypes, depending on their tissue origin, as well as on their functional state. Results In order to contribute to the proteomic characterization of fibroblasts, we have isolated primary human fibroblasts from human skin, lung and bone marrow and generated proteome profiles of these cells by LC-MS/MS. Comparative proteome profiling revealed characteristic differences therein, which seemed to be related to the cell’s tissue origin. Furthermore, the cells were treated in vitro with the pro-inflammatory cytokine IL-1beta. While all fibroblasts induced the secretion of Interleukins IL-6 and IL-8 and the chemokine GRO-alpha, other inflammation-related proteins were up-regulated in an apparently tissue-dependent manner. Investigating fibroblasts from tumorous tissues of skin, lung and bone marrow with respect to such inflammation-related proteins revealed hardly any conformity but rather individual and tumor type-related variations. However, apparent up-regulation of IGF-II, PAI-1 and PLOD2 was observed in melanoma-, lung adenocarcinoma- and multiple myeloma-associated fibroblasts, as well as in hepatocellular carcinoma-associated fibroblasts. Conclusions Inflammation-related proteome alterations of primary human fibroblasts were determined by the analysis of IL-1beta treated cells. Tumor-associated fibroblasts from different tissue types hardly showed signs of acute inflammation but displayed characteristic functional aberrations potentially related to chronic inflammation. The present data suggest that the state of the tumor microenvironment is relevant for tumor progression and targeted treatment of tumor-associated fibroblasts may support anti-cancer strategies. Electronic supplementary material The online version of this article (doi:10.1186/1559-0275-11-41) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Astrid Slany
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Vienna, Austria
| | - Anastasia Meshcheryakova
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria ; Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| | - Agnes Beer
- Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria ; Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| | - Hendrik Jan Ankersmit
- Department of Thoracic Surgery, Division of Surgery, Medical University Vienna, Vienna, Austria ; Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| | - Verena Paulitschke
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christopher Gerner
- Faculty of Chemistry, Department of Analytical Chemistry, University of Vienna, Vienna, Austria ; Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria ; Christian Doppler Laboratory for the Diagnosis and Regeneration of Cardiac and Thoracic Diseases, Medical University Vienna, Vienna, Austria
| |
Collapse
|
37
|
De Langhe E, Aznar-Lopez C, De Vooght V, Vanoirbeek JA, Luyten FP, Lories RJ. Secreted frizzled related proteins inhibit fibrosis in vitro but appear redundant in vivo. FIBROGENESIS & TISSUE REPAIR 2014; 7:14. [PMID: 25317206 PMCID: PMC4196208 DOI: 10.1186/1755-1536-7-14] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/20/2014] [Indexed: 01/24/2023]
Abstract
BACKGROUND The pathogenesis of pulmonary fibrosis remains poorly understood. The Wnt signaling pathway regulates fibrogenesis in different organs. Here, we studied the role of two extracellular Wnt antagonists, secreted frizzled-related protein-1 (SFRP1) and frizzled-related protein (FRZB) on lung fibrosis in vitro and in vivo. For this purpose, we used an alveolar epithelial cell line and a lung fibroblast cell line, and the bleomycin-induced lung fibrosis model, respectively. RESULTS During the course of bleomycin-induced lung fibrosis, Sfrp1 and Frzb expression are upregulated. Expression of Sfrp1 appears much higher than that of Frzb. In vitro, recombinant SFRP1, but not FRZB, counteracts the transforming growth factor β1 (TGFβ1)-induced upregulation of type I collagen expression both in pulmonary epithelial cells and fibroblasts. Both SFRP1 and FRZB inhibit the TGFβ1-induced increase of active β-catenin, but do not influence the TGFβ1-induced phosphorylation levels of SMAD3, positioning Wnt signaling activity downstream of the active TGFβ signal in lung fibroblasts, but not in alveolar epithelial cells. In vivo, Sfrp1 (-/-) and Frzb (-/-) mice showed identical responses to bleomycin in the lung compared to wild-type controls. CONCLUSIONS Although SFRP1 counteracts the effect of TGFβ1 in pulmonary cells in vitro; loss of neither SFRP1 nor FRZB alters fibrotic outcomes in the lungs in vivo. The lack of in vivo effect in the absence of specific SFRPs suggests functional redundancy within this family of Wnt antagonists.
Collapse
Affiliation(s)
- Ellen De Langhe
- Department of Development and Regeneration, Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium ; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Carolina Aznar-Lopez
- Department of Development and Regeneration, Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium
| | - Vanessa De Vooght
- Department of Public Health, Experimental Toxicology Unit, KU Leuven, Leuven, Belgium
| | - Jeroen A Vanoirbeek
- Department of Public Health, Experimental Toxicology Unit, KU Leuven, Leuven, Belgium
| | - Frank P Luyten
- Department of Development and Regeneration, Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium ; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| | - Rik Ju Lories
- Department of Development and Regeneration, Laboratory of Tissue Homeostasis and Disease, Skeletal Biology and Engineering Research Center, KU Leuven, Leuven, Belgium ; Division of Rheumatology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Shiomi T, Sklepkiewicz P, Bodine PVN, D'Armiento JM. Maintenance of the bronchial alveolar stem cells in an undifferentiated state by secreted frizzled-related protein 1. FASEB J 2014; 28:5242-9. [PMID: 25212222 DOI: 10.1096/fj.13-242735] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Bronchoalveolar stem cells (BASCs) are mobilized during injury and identified as lung progenitor cells, but the molecular regulation of this population of cells has not been elucidated. Secreted frizzled-related protein 1 (SFRP1) is a critical molecule involved in alveolar duct formation in the lung and here we demonstrate its importance in controlling cell differentiation during lung injury. Mice lacking SFRP1 exhibited a rapid repair response leading to aberrant proliferation of differentiated cells. Furthermore, SFRP1 treatment of BASCs maintained these cells in a quiescent state. In vivo overexpression of SFRP1 after injury suppressed differentiation and resulted in the accumulation of BASCs correlating with in vitro studies. These findings suggest that SFRP1 expression in the adult maintains progenitor cells within their undifferentiated state and suggests that manipulation of this pathway is a potential target to augment the lung repair process during disease.
Collapse
Affiliation(s)
- Takayuki Shiomi
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA; and
| | - Piotr Sklepkiewicz
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA; and
| | | | - Jeanine M D'Armiento
- Department of Anesthesiology, College of Physicians and Surgeons, Columbia University, New York, New York, USA; and
| |
Collapse
|
39
|
West JD, Austin ED, Gaskill C, Marriott S, Baskir R, Bilousova G, Jean JC, Hemnes AR, Menon S, Bloodworth NC, Fessel JP, Kropski JA, Irwin D, Ware LB, Wheeler L, Hong CC, Meyrick B, Loyd JE, Bowman AB, Ess KC, Klemm DJ, Young PP, Merryman WD, Kotton D, Majka SM. Identification of a common Wnt-associated genetic signature across multiple cell types in pulmonary arterial hypertension. Am J Physiol Cell Physiol 2014; 307:C415-30. [PMID: 24871858 PMCID: PMC4154073 DOI: 10.1152/ajpcell.00057.2014] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Accepted: 05/23/2014] [Indexed: 12/24/2022]
Abstract
Understanding differences in gene expression that increase risk for pulmonary arterial hypertension (PAH) is essential to understanding the molecular basis for disease. Previous studies on patient samples were limited by end-stage disease effects or by use of nonadherent cells, which are not ideal to model vascular cells in vivo. These studies addressed the hypothesis that pathological processes associated with PAH may be identified via a genetic signature common across multiple cell types. Expression array experiments were initially conducted to analyze cell types at different stages of vascular differentiation (mesenchymal stromal and endothelial) derived from PAH patient-specific induced pluripotent stem (iPS) cells. Molecular pathways that were altered in the PAH cell lines were then compared with those in fibroblasts from 21 patients, including those with idiopathic and heritable PAH. Wnt was identified as a target pathway and was validated in vitro using primary patient mesenchymal and endothelial cells. Taken together, our data suggest that the molecular lesions that cause PAH are present in all cell types evaluated, regardless of origin, and that stimulation of the Wnt signaling pathway was a common molecular defect in both heritable and idiopathic PAH.
Collapse
Affiliation(s)
- James D West
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Eric D Austin
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee
| | - Christa Gaskill
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Shennea Marriott
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Rubin Baskir
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| | - Ganna Bilousova
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | | | - Anna R Hemnes
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Swapna Menon
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | | | - Joshua P Fessel
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee
| | - Johnathan A Kropski
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - David Irwin
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | - Lorraine B Ware
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - Lisa Wheeler
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Charles C Hong
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Veterans Administration Hospital, Nashville, Tennessee
| | - Barbara Meyrick
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee
| | - James E Loyd
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee
| | - Aaron B Bowman
- Department of Neurology, Vanderbilt Brain Institute, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - Kevin C Ess
- Department of Pediatrics, Vanderbilt University, Nashville, Tennessee; Department of Neurology, Vanderbilt Brain Institute, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - Dwight J Klemm
- Gates Center for Regenerative Medicine and Stem Cell Biology, University of Colorado, Aurora, Colorado
| | - Pampee P Young
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | | | - Susan M Majka
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University, Nashville, Tennessee; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee; Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, Tennessee; Vanderbilt Center for Stem Cell Biology, Nashville, Tennessee; Vanderbilt Vascular Biology Center, Nashville, Tennessee; Pulmonary Vascular Research Institute, Kochi, and AnalyzeDat Consulting Services, Kerala, India; and
| |
Collapse
|
40
|
Foronjy RF, Majka SM. The potential for resident lung mesenchymal stem cells to promote functional tissue regeneration: understanding microenvironmental cues. Cells 2014; 1:874. [PMID: 23626909 PMCID: PMC3634590 DOI: 10.3390/cells1040874] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Tissue resident mesenchymal stem cells (MSCs) are important regulators of tissue repair or regeneration, fibrosis, inflammation, angiogenesis and tumor formation. Bone marrow derived mesenchymal stem cells (BM-MSCs) and endothelial progenitor cells (EPC) are currently being considered and tested in clinical trials as a potential therapy in patients with such inflammatory lung diseases including, but not limited to, chronic lung disease, pulmonary arterial hypertension (PAH), pulmonary fibrosis (PF), chronic obstructive pulmonary disease (COPD)/emphysema and asthma. However, our current understanding of tissue resident lung MSCs remains limited. This review addresses how environmental cues impact on the phenotype and function of this endogenous stem cell pool. In addition, it examines how these local factors influence the efficacy of cell-based treatments for lung diseases.
Collapse
Affiliation(s)
- Robert F. Foronjy
- Department of Medicine, St. Luke’s Roosevelt Health Sciences Center, Antenucci Building, 432 West 58th Street, Room 311, New York, NY 10019, USA; ; Tel.: +1-212-523-7265
| | - Susan M. Majka
- Department of Medicine, Vanderbilt University, 1161 21st. Ave S, T1218 MCN, Nashville, TN 37232, USA
- Author to whom correspondence should be addressed; ; Tel.: +1-303-883-8786
| |
Collapse
|
41
|
Siddiqui S, Xin Y, Emami K, D'Armiento J, Shiomi T, Profka H, Mongkolwisetwara P, Rizi R. Hyperpolarized (3)He diffusion MRI and histology of secreted frizzled related protein-1 (SFRP1) deficient lungs in a Murine model. Magn Reson Imaging 2014; 32:535-40. [PMID: 24629512 DOI: 10.1016/j.mri.2014.01.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 12/04/2013] [Accepted: 01/27/2014] [Indexed: 12/25/2022]
Abstract
Secreted frizzled related protein-1 (SFRP1) plays a key role in many diverse processes, including embryogenesis, tissue repair, bone formation, and tumor genesis. Previous studies have shown the effects of the SFRP1 gene on lung development using the SFRP1 knockout mouse model via histological and physiological studies. In this study, the feasibility of ADC (acquired via HP (3)He) to detect altered lung structure in the SFRP1 knockout (SFRP1(-/-)) mice was investigated, and compared to analysis by histology. This study consisted of two groups, the wild-type (WT) mice and the knockout (KO) mice with n=6 mice for each group. (3)He ADC MRI and histology were performed on all of the animals. The global Lm values of WT and KO mice were 35.0±0.8μm and 38.4±3.8μm, respectively, which translated to an increase of 9.58% in the Lm of KO mice. The mean global ADCs for the WT and KO mice were 0.12±0.01cm(2)/s and 0.13±0.01cm(2)/s, respectively, which equated to a relative increase of 8.0% in the KO mice compared to the WT mice. In the sub-analysis of the anterior, medial and posterior lung regions, Lm increased by 10.50%, 6.66% and 11.84% in the KO mice, respectively, whereas the differences in ADC between the two groups in the anterior, medial, and posterior regions were 7.3%, 8.3%, and 4.6%, respectively. These results suggest that HP MRI measurements can be used as a suitable substitute for histology to obtain valuable information about lung geometry non-invasively. This technique is also advantageous as regional measurements can be performed, which can identify lung destruction more precisely. Most importantly, this approach extends far beyond the specific pathology analyzed in this study, as it can be applied to many other pathological conditions in the lung tissue, as well to many other embryonic studies.
Collapse
Affiliation(s)
- Sarmad Siddiqui
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA.
| | - Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Kiarash Emami
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Takayuki Shiomi
- Department of Medicine, Columbia University, New York, NY, USA
| | - Harrilla Profka
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Rahim Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
42
|
Panaccione I, Napoletano F, Forte AM, Kotzalidis GD, Del Casale A, Rapinesi C, Brugnoli C, Serata D, Caccia F, Cuomo I, Ambrosi E, Simonetti A, Savoja V, De Chiara L, Danese E, Manfredi G, Janiri D, Motolese M, Nicoletti F, Girardi P, Sani G. Neurodevelopment in schizophrenia: the role of the wnt pathways. Curr Neuropharmacol 2013; 11:535-558. [PMID: 24403877 PMCID: PMC3763761 DOI: 10.2174/1570159x113119990037] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 03/28/2013] [Accepted: 05/12/2013] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES To review the role of Wnt pathways in the neurodevelopment of schizophrenia. METHODS SYSTEMATIC PUBMED SEARCH, USING AS KEYWORDS ALL THE TERMS RELATED TO THE WNT PATHWAYS AND CROSSING THEM WITH EACH OF THE FOLLOWING AREAS: normal neurodevelopment and physiology, neurodevelopmental theory of schizophrenia, schizophrenia, and antipsychotic drug action. RESULTS Neurodevelopmental, behavioural, genetic, and psychopharmacological data point to the possible involvement of Wnt systems, especially the canonical pathway, in the pathophysiology of schizophrenia and in the mechanism of antipsychotic drug action. The molecules most consistently found to be associated with abnormalities or in antipsychotic drug action are Akt1, glycogen synthase kinase3beta, and beta-catenin. However, the extent to which they contribute to the pathophysiology of schizophrenia or to antipsychotic action remains to be established. CONCLUSIONS The study of the involvement of Wnt pathway abnormalities in schizophrenia may help in understanding this multifaceted clinical entity; the development of Wnt-related pharmacological targets must await the collection of more data.
Collapse
Affiliation(s)
- Isabella Panaccione
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Flavia Napoletano
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Alberto Maria Forte
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Giorgio D. Kotzalidis
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Antonio Del Casale
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Chiara Rapinesi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Chiara Brugnoli
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Daniele Serata
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Federica Caccia
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Ilaria Cuomo
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Elisa Ambrosi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Alessio Simonetti
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Valeria Savoja
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Lavinia De Chiara
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Emanuela Danese
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Giovanni Manfredi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | - Delfina Janiri
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
| | | | - Ferdinando Nicoletti
- NEUROMED, Pozzilli, Isernia, Italy
- Department of Neuropharmacology, Sapienza University, School of Medicine and Pharmacy, Rome, Italy
| | - Paolo Girardi
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
| | - Gabriele Sani
- NESMOS Department (Neuroscience, Mental Health, and Sensory Organs), Sapienza University, School of Medicine and Psychology, Sant’Andrea Hospital, Rome, Italy
- Centro Lucio Bini, Rome, Italy
- IRCCS Santa Lucia Foundation, Department of Clinical and Behavioural Neurology, Neuropsychiatry Laboratory, Rome, Italy
| |
Collapse
|
43
|
Abstract
Increasing protein expression enables researchers to better understand the functional role of that protein in regulating key biological processes(1). In the lung, this has been achieved typically through genetic approaches that utilize transgenic mice(2,3) or viral or non-viral vectors that elevate protein levels via increased gene expression(4). Transgenic mice are costly and time-consuming to generate and the random insertion of a transgene or chronic gene expression can alter normal lung development and thus limit the utility of the model(5). While conditional transgenics avert problems associated with chronic gene expression(6), the reverse tetracycline-controlled transactivator (rtTA) mice, which are used to generate conditional expression, develop spontaneous air space enlargement(7). As with transgenics, the use of viral and non-viral vectors is expensive(8) and can provoke dose-dependent inflammatory responses that confound results(9) and hinder expression(10). Moreover, the efficacy of repeated doses are limited by enhanced immune responses to the vector(11,12). Researchers are developing adeno-associated viral (AAV) vectors that provoke less inflammation and have longer expression within the lung(13). Using β-galactosidase, we present a method for rapidly and effectively increasing protein expression within the lung using a direct protein transfection technique. This protocol mixes a fixed amount of purified protein with 20 μl of a lipid-based transfection reagent (Pro-Ject, Pierce Bio) to allow penetration into the lung tissue itself. The liposomal protein mixture is then injected into the lungs of the mice via the trachea using a microsprayer (Penn Century, Philadelphia, PA). The microsprayer generates a fine plume of liquid aerosol throughout the lungs. Using the technique we have demonstrated uniform deposition of the injected protein throughout the airways and the alveoli of mice(14). The lipid transfection technique allows the use of a small amount of protein to achieve effect. This limits the inflammatory response that otherwise would be provoked by high protein administration. Indeed, using this technique we published that we were able to significantly increase PP2A activity in the lung without affecting lung lavage cellularity(15). Lung lavage cellularity taken 24 hr after challenge was comparable to controls (27 ± 4 control vs. 31 ± 5 albumin transfected; N=6 per group). Moreover, it increases protein levels without inducing lung developmental changes or architectural changes that can occur in transgenic models. However, the need for repeated administrations may make this technique less favorable for studies examining the effects of long-term increases in protein expression. This would be particularly true for proteins with short half-lives.
Collapse
|
44
|
Chow K, Fessel JP, Kaoriihida-Stansbury, Schmidt EP, Gaskill C, Alvarez D, Graham B, Harrison DG, Wagner DH, Nozik-Grayck E, West JD, Klemm DJ, Majka SM. Dysfunctional resident lung mesenchymal stem cells contribute to pulmonary microvascular remodeling. Pulm Circ 2013; 3:31-49. [PMID: 23662173 PMCID: PMC3641738 DOI: 10.4103/2045-8932.109912] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pulmonary vascular remodeling and oxidative stress are common to many adult lung diseases. However, little is known about the relevance of lung mesenchymal stem cells (MSCs) in these processes. We tested the hypothesis that dysfunctional lung MSCs directly participate in remodeling of the microcirculation. We employed a genetic model to deplete extracellular superoxide dismutase (EC-SOD) in lung MSCs coupled with lineage tracing analysis. We crossed floxpsod3 and mT/mG reporter mice to a strain expressing Cre recombinase under the control of the ABCG2 promoter. We demonstrated In vivo that depletion of EC-SOD in lung MSCs resulted in their contribution to microvascular remodeling in the smooth muscle actin positive layer. We further characterized lung MSCs to be multipotent vascular precursors, capable of myofibroblast, endothelial and pericyte differentiation in vitro. EC-SOD deficiency in cultured lung MSCs accelerated proliferation and apoptosis, restricted colony-forming ability, multilineage differentiation potential and promoted the transition to a contractile phenotype. Further studies correlated cell dysfunction to alterations in canonical Wnt/β-catenin signaling, which were more evident under conditions of oxidative stress. Our data establish that lung MSCs are a multipotent vascular precursor population, a population which has the capacity to participate in vascular remodeling and their function is likely regulated in part by the Wnt/β-catenin signaling pathway. These studies highlight an important role for microenviromental regulation of multipotent MSC function as well as their potential to contribute to tissue remodeling.
Collapse
Affiliation(s)
- Kelsey Chow
- Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
The WNT signaling pathway from ligand secretion to gene transcription: molecular mechanisms and pharmacological targets. Pharmacol Ther 2013; 138:66-83. [PMID: 23328704 DOI: 10.1016/j.pharmthera.2013.01.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 12/20/2012] [Indexed: 12/22/2022]
Abstract
Wingless/integrase-1 (WNT) signaling is a key pathway regulating various aspects of embryonic development; however it also underlies several pathological conditions in man, including various cancers and fibroproliferative diseases in several organs. Investigating the molecular processes involved in (canonical) WNT signaling will open new avenues for generating new therapeutics to specifically target diseases in which WNT signaling is aberrantly regulated. Here we describe the complexity of WNT signal transduction starting from the processes involved in WNT ligand biogenesis and secretion by WNT producing cells followed by a comprehensive overview of the molecular signaling events ultimately resulting in enhanced transcription of specific genes in WNT receiving cells. Finally, the possible targets for therapeutic intervention and the available pharmacological inhibitors for this complex signaling pathway are discussed.
Collapse
|
46
|
Boucherat O, Chakir J, Jeannotte L. The loss of Hoxa5 function promotes Notch-dependent goblet cell metaplasia in lung airways. Biol Open 2012; 1:677-91. [PMID: 23213461 PMCID: PMC3507293 DOI: 10.1242/bio.20121701] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Hox genes encode transcription factors controlling complex developmental processes in various organs. Little is known, however, about how HOX proteins control cell fate. Herein, we demonstrate that the goblet cell metaplasia observed in lung airways from Hoxa5−/− mice originates from the transdifferentiation of Clara cells. Reduced CC10 expression in Hoxa5−/− embryos indicates that altered cell specification occurs prior to birth. The loss of Hoxa5 function does not preclude airway repair after naphthalene exposure, but the regenerated epithelium presents goblet cell metaplasia and less CC10-positive cells, demonstrating the essential role of Hoxa5 for correct differentiation. Goblet cell metaplasia in Hoxa5−/− mice is a FOXA2-independent process. However, it is associated with increased Notch signaling activity. Consistent with these findings, expression levels of activated NOTCH1 and the effector gene HEY2 are enhanced in patients with chronic obstructive pulmonary disease. In vivo administration of a γ-secretase inhibitor attenuates goblet cell metaplasia in Hoxa5−/− mice, highlighting the contribution of Notch signaling to the phenotype and suggesting a potential therapeutic strategy to inhibit goblet cell differentiation and mucus overproduction in airway diseases. In summary, the loss of Hoxa5 function in lung mesenchyme impacts on epithelial cell fate by modulating Notch signaling.
Collapse
Affiliation(s)
- Olivier Boucherat
- Centre de recherche en cancérologie de l'Université Laval, Centre Hospitalier Universitaire de Québec , L'Hôtel-Dieu de Québec, 9 rue McMahon, Québec QC G1R 2J6 , Canada
| | | | | |
Collapse
|
47
|
Jun D, Garat C, West J, Thorn N, Chow K, Cleaver T, Sullivan T, Torchia EC, Childs C, Shade T, Tadjali M, Lara A, Nozik-Grayck E, Malkoski S, Sorrentino B, Meyrick B, Klemm D, Rojas M, Wagner DH, Majka SM. The pathology of bleomycin-induced fibrosis is associated with loss of resident lung mesenchymal stem cells that regulate effector T-cell proliferation. Stem Cells 2011; 29:725-35. [PMID: 21312316 PMCID: PMC3322548 DOI: 10.1002/stem.604] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Tissue-resident mesenchymal stem cells (MSCs) are important regulators of tissue repair or regeneration, fibrosis, inflammation, angiogenesis, and tumor formation. Here, we define a population of resident lung MSCs (luMSCs) that function to regulate the severity of bleomycin injury via modulation of the T-cell response. Bleomycin-induced loss of these endogenous luMSCs and elicited fibrosis (pulmonary fibrosis), inflammation, and pulmonary arterial hypertension (PAH). Replacement of resident stem cells by administration of isolated luMSCs attenuated the bleomycin-associated pathology and mitigated the development of PAH. In addition, luMSC modulated a decrease in numbers of lymphocytes and granulocytes in bronchoalveolar fluid and demonstrated an inhibition of effector T-cell proliferation in vitro. Global gene expression analysis indicated that the luMSCs are a unique stromal population differing from lung fibroblasts in terms of proinflammatory mediators and profibrotic pathways. Our results demonstrate that luMSCs function to protect lung integrity after injury; however, when endogenous MSCs are lost, this function is compromised illustrating the importance of this novel population during lung injury. The definition of this population in vivo in both murine and human pulmonary tissue facilitates the development of a therapeutic strategy directed at the rescue of endogenous cells to facilitate lung repair during injury.
Collapse
Affiliation(s)
- Du Jun
- Charles C. Gates Center for Regenerative Medicine and Stem Cell Biology Program, University of Colorado Denver, Aurora, Colorado 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kneidinger N, Yildirim AÖ, Callegari J, Takenaka S, Stein MM, Dumitrascu R, Bohla A, Bracke KR, Morty RE, Brusselle GG, Schermuly RT, Eickelberg O, Königshoff M. Activation of the WNT/β-catenin pathway attenuates experimental emphysema. Am J Respir Crit Care Med 2010; 183:723-33. [PMID: 20889911 DOI: 10.1164/rccm.200910-1560oc] [Citation(s) in RCA: 143] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Chronic obstructive pulmonary disease (COPD) is a devastating disease, for which no causal therapy is available. OBJECTIVES To characterize WNT/β-catenin signaling in COPD in humans and elucidate its potential role as a preventive and therapeutic target in experimental emphysema in mice. METHODS The expression, localization, and activity of WNT/β-catenin signaling was assessed in 12 COPD and 12 transplant donor samples using quantitative reverse transcriptase polymerase chain reaction, immunohistochemistry, and Western blotting. The role of WNT/β-catenin signaling was assessed in elastase- and cigarette smoke-induced emphysema and therapeutic modulation thereof in elastase-induced emphysema in TOPGAL reporter and wild-type mice in vivo. MEASUREMENTS AND MAIN RESULTS No differences in the mRNA expression profile of the main WNT/β-catenin signaling components were observed comparing COPD and donor lung homogenates. Immunohistochemical analysis revealed reduced numbers of nuclear β-catenin-positive alveolar epithelial cells in COPD. Similarly, WNT/β-catenin signaling was down-regulated in both experimental emphysema models. Preventive and therapeutic, WNT/β-catenin activation by lithium chloride attenuated experimental emphysema, as assessed by decreased airspace enlargement, improved lung function, reduced collagen content, and elevated expression of alveolar epithelial cell markers. CONCLUSIONS Decreased WNT/β-catenin signaling is involved in parenchymal tissue destruction and impaired repair capacity in emphysema. These data indicate a crucial role of WNT/β-catenin signaling in lung repair mechanisms in vivo, and highlight WNT/β-catenin activation as a future therapeutic approach for emphysema.
Collapse
Affiliation(s)
- Nikolaus Kneidinger
- Department of Medicine, University of Giessen Lung Center, University of Giessen, Giessen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|