1
|
Gillesberg FS, Pehrsson M, Bay-Jensen AC, Frederiksen P, Karsdal M, Deleuran BW, Kragstrup TW, Kubo S, Tanaka Y, Mortensen JH. Regulation of fibronectin and collagens type I, III and VI by TNF-α, TGF-β, IL-13, and tofacitinib. Sci Rep 2025; 15:1087. [PMID: 39774197 PMCID: PMC11707072 DOI: 10.1038/s41598-024-84151-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Understanding how inflammatory cytokines influence profibrogenic wound healing responses in fibroblasts is important for understanding the pathogenesis of fibrosis. TNF-α and IL-13 are key cytokines in Th1 and Th2 immune responses, respectively, while TGF-β1 is the principal pro-fibrotic mediator. We show that 12-day fibroblast culture with TNF-α or IL-13 induces fibrogenesis, marked by progressively increasing type III and VI collagen formation, and that TGF-β1 co-stimulation amplifies these effects. Tofacitinib substantially reduced the formation of ECM proteins in response to IL-13, while fibrogenesis in response to TNF-α or TGF-β1 was marginally inhibited. The in vitro findings were supported by clinical observations in patients with active rheumatoid arthritis, which had elevated serum type III collagen formation, indicating ongoing fibrogenesis during inflammation. After 48-60 weeks of tofacitinib treatment, type III collagen degradation, aswell as formation, were significantly decreased compared to baseline, highlighting dual anti-inflammatory and anti-fibrogenic effects of tofacitinib. In contrast, other anti-inflammatory treatments including methotrexate, adalimumab and tocilizumab demonstrated anti-inflammatory effects only. Our results highlight fibro-inflammatory profiles associated with TNF-α or IL-13 stimulation, both alone and in combination with TGF-β1, and support the use of tofacitinib as an anti-fibrogenic treatment in chronic inflammatory conditions.
Collapse
Affiliation(s)
- Frederik S Gillesberg
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark.
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark.
| | - Martin Pehrsson
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | | | - Peder Frederiksen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| | - Bent W Deleuran
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
| | - Tue W Kragstrup
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, Aarhus C, 8000, Denmark
- Department of Rheumatology, Århus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, 8200, Denmark
- Diagnostic Center, Regional Hospital Silkeborg, Falkevej 1, Silkeborg, 8600, Denmark
| | - Satoshi Kubo
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational & Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi, 807-8555, Kitakyushu, Japan
| | - Joachim H Mortensen
- Nordic Bioscience, Immunoscience, Herlev Hovedgade 205-207, Herlev, 2730, Denmark
| |
Collapse
|
2
|
Li W, Li Z, Zou Z, Liu X, Li X. Integrated single-cell and bulk RNA sequencing identifies POSTN as a potential biomarker and therapeutic target for rheumatoid arthritis. Gene 2024; 928:148798. [PMID: 39067546 DOI: 10.1016/j.gene.2024.148798] [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: 04/30/2024] [Revised: 07/03/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND This study aimed to integrate single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data to identify potential biomarkers and therapeutic targets for rheumatoid arthritis (RA). METHOD Firstly, we obtained the synovial scRNA-seq data from the Immport database and bulk RNA-seq data from the Gene Expression Omnibus (GEO) database. Then, we used weighted gene correlation network analysis (WGCNA) to screen for module genes most relevant to RA and intersected them with the differentially expressed genes (DEGs) obtained from scRNA-seq and bulk RNA-seq to obtain intersecting genes. Next, we constructed a protein-protein interaction (PPI) network of hub genes using the STRING database and Cytoscape software and validated its expression using external validation cohorts. Finally, we performed immune cell infiltration analysis using CIBERSORT and explored the expression and drug binding activity of key gene using clinical samples and molecular docking, respectively. RESULT We identified six cellular subgroups through dimensionality reduction and clustering, and fibroblasts may be the most important cell cluster in RA based on pseudotime and cell-cell communication analyses. Subsequently, we intersected module genes with DEGs obtained from scRNA-seq and bulk RNA-seq and constructed a PPI network of hub genes (BGN, COL11A1, COL1A1, GUCY1A1, POSTN). In external validation cohorts, POSTN was highly expressed and demonstrated the highest diagnostic performance (AUC = 0.716). In subsequent analyses, we defined POSTN as a key gene and found that its expression level was positively correlated with M2 macrophages in immune cell infiltration analysis. Additionally, POSTN was upregulated in clinical samples and exhibited favorable binding activity with nine anti-rheumatoid arthritis drugs (affinity ≤ -6.0 kcal/mol). CONCLUSION Through bioinformatics analysis, clinical sample validation, and molecular docking, we found that POSTN was highly expressed in RA and stably bound to common anti-rheumatoid arthritis drugs, which will provide new insights into potential biomarkers and therapeutic targets for RA.
Collapse
Affiliation(s)
- Weihua Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zhiqiang Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Zehui Zou
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xuqiang Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| | - Xiaofeng Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China; Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
3
|
Groen SS, Bay-Jensen AC, Thudium CS, Dziegiel MH, Skougaard M, Thomsen SF, Nielsen SH. Evaluating the inhibition of IL-17A and TNFα in a cartilage explant model cultured with Th17-derived cytokines. J Transl Autoimmun 2024; 8:100231. [PMID: 38292069 PMCID: PMC10826309 DOI: 10.1016/j.jtauto.2024.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Introduction T-helper 17 (Th17) cells produce IL-17A playing a critical role in activating the pathogenic chain leading to joint tissue inflammation and destruction. Elevated levels of Th17 cells and IL-17A have been detected in skin lesions, blood, and synovial fluid from patients with psoriatic arthritis (PsA) and ankylosing spondylitis (AS). Moreover, IL-17A inhibitors suppress disease activity in psoriasis, PsA and AS, supporting the evidence of IL-17A contributing to the disease pathogenesis. Although, IL-17A inhibitors are widely approved, it remains unclear how the inhibitory effect of IL-17A alters the extracellular matrix (ECM) of the joint in a Th17-conditioned inflammatory milieu. Therefore, the aim of this study was to establish a cartilage model cultured with conditioned medium from Th17 cells and inhibitors to explore the effect of IL-17A inhibition on joint tissue remodeling. Methods Naïve CD4+ T cells from healthy human buffy coat were differentiated into Th17 cells, followed by Th17 cell activation to secrete Th17-related cytokines and molecules into media. The activated Th17 cells were isolated from the conditioned media (CM) and analyzed using flow cytometry to verify Th17 cell differentiation. The CM were assessed with ELISA to quantify the concentrations of cytokines secreted into the media by the Th17 cells. Healthy bovine cartilage explants were cultured with the Th17-CM and treated with IL-17A and TNFα inhibitors for 21 days. In harvested supernatant from the cartilage cultures, MMP- and ADAMTS-mediated biomarker fragments of type II collagen, aggrecan, and fibronectin were measured by ELISA to investigate the ECM remodeling within the cartilage tissue. Results Th17-CM stimulated a catabolic response in the cartilage. Markers of type II collagen and aggrecan degradation were upregulated, while anabolic marker of type II collagen formation remained on similar levels as the untreated explants. The addition of IL-17A inhibitor to Th17-CM decreased the elevated type II collagen and aggrecan degradation, however, degenerative levels were still elevated compared to untreated group. The addition of TNFα inhibitor completely reduced both type II collagen and aggrecan degradation compared to untreated explants. Moreover, the TNFα inhibitor treatment did not alter the type II collagen formation compared to untreated group. Conclusion This study suggests that inhibition of IL-17A in Th17-conditioned cartilage tissue only partially reduced the MMP-mediated type II collagen degradation and ADAMTS-mediated aggrecan degradation, while the TNFα inhibitor treatment fully reduced both MMP- and ADAMTS-mediated ECM degradation. This exploratory study where ECM biomarkers are combined with Th17-conditioned ex vivo model may hold great potential as output for describing joint disease mechanisms and predicting structural effects of treatment on joint tissue.
Collapse
Affiliation(s)
- Solveig Skovlund Groen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Morten H. Dziegiel
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Marie Skougaard
- The Copenhagen Center for Translational Research, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- The Parker Institute, Bispebjerg and Frederiksberg Hospital, Frederiksberg, Denmark
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | - Simon Francis Thomsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Dermatology, Bispebjerg Hospital, Copenhagen, Denmark
| | - Signe Holm Nielsen
- Immunoscience, Nordic Bioscience, Herlev, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| |
Collapse
|
4
|
Groen SS, Nielsen SH, Bay-Jensen AC, Rasti M, Ganatra D, Oikonomopoulou K, Chandran V. Investigating protease-mediated peptides of inflammation and tissue remodeling as biomarkers associated with flares in psoriatic arthritis. Arthritis Res Ther 2024; 26:107. [PMID: 38802975 PMCID: PMC11129460 DOI: 10.1186/s13075-024-03332-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Psoriatic arthritis (PsA) is an inflammatory arthritis associated with psoriasis. PsA disease involves flares, which are associated with increased joint inflammation and tissue remodeling. There is a need for identifying biomarkers related to PsA disease activity and flares to improve the management of PsA patients and decrease flares. The tissue turnover imbalance that occurs during the inflammatory and fibro-proliferative processes during flares leads to an increased degradation and/or reorganization of the extracellular matrix (ECM), where increased proteolysis plays a key role. Hence, protease-mediated fragments of inflammatory and tissue-remodeling components could be used as markers reflecting flares in PsA patients. METHODS A broad panel of protease-mediated biomarkers reflecting inflammation and tissue remodeling was measured in serum and synovial fluid (SF) obtained from PsA patients experiencing flares (acutely swollen joint[s], PsA-flare). In serum, biomarker levels assessed in PsA-flare patients were compared to controls and in early-diagnosed PsA patients not experiencing flares (referred to as PsA without flare). Furthermore, the biomarker levels assessed in SF from PsA-flare patients were compared to the levels in SF of osteoarthritis (OA) patients. RESULTS In serum, levels of the PRO-C3 and C3M, reflecting formation and degradation of the interstitial matrix, were found significantly elevated in PsA-flare compared to controls and PsA without flare. The remodeling marker of the basement membrane, PRO-C4, was significantly elevated in PsA-flare compared to PsA without flare. The inflammation and immune cell activity related markers, CRPM, VICM, and CPa9-HNE were significantly elevated in PsA-flare patients compared to controls and PsA without flare. In addition, VICM (AUC = 0.71), CPa9-HNE (AUC = 0.89), CRPM (AUC = 0.76), and PRO-C3 (AUC = 0.86) showed good discriminatory performance for separating PsA-flare from PsA without flare. In SF, the macrophage activity marker, VICM, was significantly elevated whereas the type II collagen formation marker, PRO-C2, was significantly reduced in the PsA-flare compared to OA. The combination of five serum markers reflecting type III and IV collagen degradation (C3M and C4M, respectively), type III and VI collagen formation (PRO-C3 and PRO-C6, respectively), and neutrophil activity (CPa9-HNE) showed an excellent discriminatory performance (AUC = 0.98) for separating PsA-flare from PsA without flares. CONCLUSIONS The serum biomarker panel of C3M, C4M, PRO-C3, PRO-C6, and CPa9-HNE reflecting synovitis, enthesitis, and neutrophil activity may serve as novel tool for quantitatively monitoring flares in PsA patients.
Collapse
Affiliation(s)
- Solveig Skovlund Groen
- ImmunoScience, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark.
- Biomedical Sciences, University of Copenhagen, Copenhagen N, Denmark.
| | - Signe Holm Nielsen
- ImmunoScience, Biomarkers and Research, Nordic Bioscience, Herlev, Denmark
- Biotechnology and Biomedicine, Technical University of Denmark, Kgs. Lyngby, Denmark
| | | | - Mozhgan Rasti
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Darshini Ganatra
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Katerina Oikonomopoulou
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Vinod Chandran
- Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Medicine, Division of Rheumatology, Department of Laboratory Medicine and Pathobiology, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Fan Z, Chen F, Liu Y, Huang X, Tian S, Ma Y. Expression and Clinicopathological Significance of Extracellular Matrix Remodeling Markers in Esophageal Squamous Carcinoma. Crit Rev Eukaryot Gene Expr 2024; 34:71-78. [PMID: 38912964 DOI: 10.1615/critreveukaryotgeneexpr.2024053646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/25/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignancy of the gastrointestinal tract with a single therapeutic option and a lack of effective clinical therapeutic biomarkers. Extracellular matrix (ECM) remodeling plays a pro-carcinogenic role in a variety of malignancies, but its role in esophageal squamous carcinoma remains to be elucidated. In this study, we examined the expression levels of ECM remodeling markers in 71 pairs of esophageal squamous carcinoma tissues and normal tissues adjacent to the carcinoma using immunohistochemical staining, and analyzed their relationship with clinicopathological features and prognosis. The results suggested that extracellular matrix remodeling markers (integrin αV, fibronectin, MMP9) were abnormally highly expressed in esophageal squamous carcinoma tissues. There was a statistically significant difference between the positive expression of ECM remodeling and the TNM stage of esophageal squamous carcinoma, and there was no statistically significant correlation with age, gender and carcinoembryonic antigen expression, differentiation degree, T stage, and lymph node metastasis. Overall survival rate and overall survival time were significantly lower in patients with positive ECM remodeling expression, which was an independent risk factor for poor prognosisof esophageal squamous carcinoma.
Collapse
Affiliation(s)
- Zhiqin Fan
- Department of Pathology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China; Department of Daily Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
| | - Fei Chen
- Department of Daily Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
| | - Yingmin Liu
- Department of Daily Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
| | - Xiaotong Huang
- Department of Daily Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
| | - Siyue Tian
- Department of Daily Surgery, Affiliated Tumor Hospital, Xinjiang Medical University, Urumqi 830011, Xinjiang Uygur Autonomous Region, China
| | - Yuqing Ma
- Department of Pathology, The First Affiliated Hospital, Xinjiang Medical University, Urumqi 830054, Xinjiang Uygur Autonomous Region, China
| |
Collapse
|
6
|
Hasselbalch HC, Junker P, Skov V, Kjær L, Knudsen TA, Larsen MK, Holmström MO, Andersen MH, Jensen C, Karsdal MA, Willumsen N. Revisiting Circulating Extracellular Matrix Fragments as Disease Markers in Myelofibrosis and Related Neoplasms. Cancers (Basel) 2023; 15:4323. [PMID: 37686599 PMCID: PMC10486581 DOI: 10.3390/cancers15174323] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/04/2023] [Accepted: 08/07/2023] [Indexed: 09/10/2023] Open
Abstract
Philadelphia chromosome-negative chronic myeloproliferative neoplasms (MPNs) arise due to acquired somatic driver mutations in stem cells and develop over 10-30 years from the earliest cancer stages (essential thrombocythemia, polycythemia vera) towards the advanced myelofibrosis stage with bone marrow failure. The JAK2V617F mutation is the most prevalent driver mutation. Chronic inflammation is considered to be a major pathogenetic player, both as a trigger of MPN development and as a driver of disease progression. Chronic inflammation in MPNs is characterized by persistent connective tissue remodeling, which leads to organ dysfunction and ultimately, organ failure, due to excessive accumulation of extracellular matrix (ECM). Considering that MPNs are acquired clonal stem cell diseases developing in an inflammatory microenvironment in which the hematopoietic cell populations are progressively replaced by stromal proliferation-"a wound that never heals"-we herein aim to provide a comprehensive review of previous promising research in the field of circulating ECM fragments in the diagnosis, treatment and monitoring of MPNs. We address the rationales and highlight new perspectives for the use of circulating ECM protein fragments as biologically plausible, noninvasive disease markers in the management of MPNs.
Collapse
Affiliation(s)
- Hans Carl Hasselbalch
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Peter Junker
- Department of Rheumatology, Odense University Hospital, 5000 Odense, Denmark;
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Trine A. Knudsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Kranker Larsen
- Department of Hematology, Zealand University Hospital, 4000 Roskilde, Denmark; (V.S.); (L.K.); (T.A.K.); (M.K.L.)
| | - Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, 2730 Herlev, Denmark; (M.O.H.); (M.H.A.)
| | - Christina Jensen
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | - Morten A. Karsdal
- Nordic Bioscience A/S, 2730 Herlev, Denmark; (C.J.); (M.A.K.); (N.W.)
| | | |
Collapse
|
7
|
Madsen SF, Sand JMB, Juhl P, Karsdal M, Thudium CS, Siebuhr AS, Bay-Jensen AC. Fibroblasts are not just fibroblasts: clear differences between dermal and pulmonary fibroblasts' response to fibrotic growth factors. Sci Rep 2023; 13:9411. [PMID: 37296166 PMCID: PMC10256773 DOI: 10.1038/s41598-023-36416-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Systemic Sclerosis (SSc) hallmark is skin fibrosis, but up to 80% of the patients have fibrotic involvement in the pulmonary system. Antifibrotic drugs which have failed in a general SSc population have now been approved in patients with SSc-associated interstitial lung disease (ILD). This indicates that the fibrotic progression and regulation of fibroblasts likely depend on local factors specific to the tissue type. This study investigated the difference between dermal and pulmonary fibroblasts in a fibrotic setting, mimicking the extracellular matrix. Primary healthy fibroblasts were grown in a crowded environment and stimulated with TGF-β1 and PDGF-AB. The viability, morphology, migration capacity, extracellular matrix formation, and gene expression were assessed: TGF-β1 only increased the viability in the dermal fibroblasts. PDGF-AB increased the migration capacity of dermal fibroblasts while the pulmonary fibroblasts fully migrated. The morphology of the fibroblasts was different without stimulation. TGF-β1 increased the formation of type III collagen in pulmonary fibroblasts, while PDGF-AB increased it in dermal fibroblasts. The gene expression trend of type VI collagen was the opposite after PDGF-AB stimulation. The fibroblasts exhibit different response profiles to TGF-β1 and PDGF-AB; this suggests that drivers of fibrosis are tissue-dependent, which needs to be considered in drug development.
Collapse
Affiliation(s)
- Sofie Falkenløve Madsen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Immunoscience, Nordic Bioscience, Herlev, Denmark.
| | | | | | | | | | | | | |
Collapse
|
8
|
Zhang Y, Feng Y, Shao Q, Jiang Z, Yang G. Rapid formation of 3D: Decellularized extracellular matrix spheroids for enhancing bone formation. J Biomed Mater Res A 2023; 111:378-388. [PMID: 36355784 DOI: 10.1002/jbm.a.37471] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/29/2022] [Accepted: 10/31/2022] [Indexed: 11/12/2022]
Abstract
Bone marrow mesenchymal stem cell sheet-derived spheroids (BMSCs spheroids) have been widely studied as native bioactive scaffolds. However, the abundant cells in BMSCs spheroids cause immunogenicity and make them difficult to store. This paper aimed to construct a new bioactive scaffold called 3D-decellularized extracellular matrix spheroids (ECM spheroids) via decellularization of BMSCs spheroids to enhance bone formation. Hematoxylin and eosin staining (HE), nuclear and cytoskeletal fluorescence, immunofluorescence (IF), and scanning electron microscopy (SEM) were utilized to detect the characteristics and components of ECM spheroids. Furthermore, the biological properties of migration, adhesion, and recellularization of cells in ECM spheroids were assessed in vitro, and bone formation was evaluated in rat calvarial defects. The results showed that both the nuclei and cytoskeleton in ECM spheroids were greatly altered and one of the major components of FN was intact. The migration, adhesion, and recellularization potential were improved in vitro. Meanwhile, ECM spheroids promoted osteogenesis in rat skull defects after 3 months (p < .01). In conclusion, ECM spheroids were successfully prepared and proven to promote cell migration, adhesion, and proliferation. Bone formation in vivo was also accelerated. We believe that ECM spheroids can be used as bioactive and biocompatible 3D scaffolds in the future.
Collapse
Affiliation(s)
- Yanmin Zhang
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou, China
| | - Yuting Feng
- Department of Preventive Dentistry, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China
| | - Qin Shao
- Department of Stomatology, Integrated Traditional and Western Medicine Hospital of Linping District, Hangzhou, China
| | - Zhiwei Jiang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China.,Department of Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China
| | - Guoli Yang
- Key Laboratory of Oral Biomedical Research of Zhejiang Province, Hangzhou, China.,Department of Implantology, Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Zhejiang, Hangzhou, China
| |
Collapse
|
9
|
Hesse C, Beneke V, Konzok S, Diefenbach C, Bülow Sand JM, Rønnow SR, Karsdal MA, Jonigk D, Sewald K, Braun A, Leeming DJ, Wollin L. Nintedanib modulates type III collagen turnover in viable precision-cut lung slices from bleomycin-treated rats and patients with pulmonary fibrosis. Respir Res 2022; 23:201. [PMID: 35927669 PMCID: PMC9351157 DOI: 10.1186/s12931-022-02116-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 07/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aberrant extracellular matrix (ECM) deposition and remodelling is important in the disease pathogenesis of pulmonary fibrosis (PF). We characterised neoepitope biomarkers released by ECM turnover in lung tissue from bleomycin-treated rats and patients with PF and analysed the effects of two antifibrotic drugs: nintedanib and pirfenidone. METHODS Precision-cut lung slices (PCLS) were prepared from bleomycin-treated rats or patients with PF. PCLS were incubated with nintedanib or pirfenidone for 48 h, and levels of neoepitope biomarkers of type I, III and VI collagen formation or degradation (PRO-C1, PRO-C3, PRO-C6 and C3M) as well as fibronectin (FBN-C) were assessed in the culture supernatants. RESULTS In rat PCLS, incubation with nintedanib led to a reduction in C3M, reflecting type III collagen degradation. In patient PCLS, incubation with nintedanib reduced the levels of PRO-C3 and C3M, thus showing effects on both formation and degradation of type III collagen. Incubation with pirfenidone had a marginal effect on PRO-C3. There were no other notable effects of either nintedanib or pirfenidone on the other neoepitope biomarkers studied. CONCLUSIONS This study demonstrated that nintedanib modulates neoepitope biomarkers of type III collagen turnover and indicated that C3M is a promising translational neoepitope biomarker of PF in terms of therapy assessment.
Collapse
Affiliation(s)
- Christina Hesse
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of German Center for Lung Research (DZL), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Valerie Beneke
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of German Center for Lung Research (DZL), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Sebastian Konzok
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of German Center for Lung Research (DZL), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Claudia Diefenbach
- Translational Medicine + Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | | | | | | | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of German Center for Lung Research (DZL), Hannover, Germany
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of German Center for Lung Research (DZL), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Member of German Center for Lung Research (DZL), Hannover, Germany.,Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of Fraunhofer International Consortium for Anti-Infective Research (iCAIR), Member of Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Hannover, Germany
| | | | - Lutz Wollin
- Translational Medicine + Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany.
| |
Collapse
|
10
|
Søndergaard RH, Højgaard LD, Reese-Petersen AL, Hoeeg C, Mathiasen AB, Haack-Sørensen M, Follin B, Genovese F, Kastrup J, Juhl M, Ekblond A. Adipose-derived stromal cells increase the formation of collagens through paracrine and juxtacrine mechanisms in a fibroblast co-culture model utilizing macromolecular crowding. Stem Cell Res Ther 2022; 13:250. [PMID: 35690799 PMCID: PMC9188050 DOI: 10.1186/s13287-022-02923-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 05/29/2022] [Indexed: 11/24/2022] Open
Abstract
Background Adipose-derived stromal cells (ASCs) possess a multitude of regenerative capabilities, which include immunomodulation, angiogenesis, and stimulation of extracellular matrix (ECM) remodeling. However, the underlying mechanisms leading to ECM remodeling remain largely elusive and highlight the need for functional in vitro models for mode of action studies. Therefore, the purpose of this study was to develop an in vitro co-culture model to investigate the capabilities of ASCs to modulate fibroblasts and ECM. Methods An ECM in vitro model with ASCs and normal human dermal fibroblasts (NHDFs) was established utilizing macromolecular crowding, ascorbic acid, and TGF-β stimulation. Paracrine and juxtacrine co-cultures were created using transwell inserts and cell cultures with direct cell–cell contacts. The cultures were screened using RT2 PCR Profiler Arrays; the protein levels of myofibroblast differentiation marker alpha smooth muscle actin (αSMA) and ECM remodeling enzymes were analyzed using western blot on cell lysates; the formation of collagen type I, III, VI, and fibronectin was investigated using ELISA on culture supernatants; and the deposition of collagens was analyzed using immunocytochemistry. Results TGF-β stimulation of NHDF monocultures increased the expression of 18 transcripts relevant for ECM formation and remodeling, the protein levels of αSMA and matrix metalloproteinase-2 (MMP-2), the formation of collagen type I, III, VI, and fibronectin, and the deposition of collagen type I and VI and decreased the protein levels of MMP-14. Inclusion of ASCs in the ECM co-culture model increased the formation of collagen type I and III through paracrine mechanisms and the formation of collagen type VI through juxtacrine mechanisms. Conclusions The co-culture model provides effective stimulation of NHDF monocultures by TGF-β for enhanced formation and deposition of ECM. In the model, ASCs induce changes in ECM by increasing formation of collagen type I, III and VI. The obtained results could guide further investigations of ASCs’ capabilities and underlying mechanisms related to ECM formation and remodeling. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02923-y.
Collapse
Affiliation(s)
- Rebekka Harary Søndergaard
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark.
| | - Lisbeth Drozd Højgaard
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | | | - Cecilie Hoeeg
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Anders Bruun Mathiasen
- Department of Cardiology, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Mandana Haack-Sørensen
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Bjarke Follin
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Federica Genovese
- Nordic Bioscience A/S, Herlev Hovedgade 205-207, 2730, Herlev, Denmark
| | - Jens Kastrup
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Morten Juhl
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| | - Annette Ekblond
- Cardiology Stem Cell Centre, The Heart Centre, Copenhagen University Hospital Rigshospitalet, Henrik Harpestrengs vej 4C, Dept. 9302, 2100, Copenhagen, Denmark
| |
Collapse
|
11
|
Decato BE, Leeming DJ, Sand JMB, Fischer A, Du S, Palmer SM, Karsdal M, Luo Y, Minnich A. LPA 1 antagonist BMS-986020 changes collagen dynamics and exerts antifibrotic effects in vitro and in patients with idiopathic pulmonary fibrosis. Respir Res 2022; 23:61. [PMID: 35303880 PMCID: PMC8933988 DOI: 10.1186/s12931-022-01980-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a debilitating lung disease with limited treatment options. A phase 2 trial (NCT01766817) showed that twice-daily treatment with BMS-986020, a lysophosphatidic acid receptor 1 (LPA1) antagonist, significantly decreased the slope of forced vital capacity (FVC) decline over 26 weeks compared with placebo in patients with IPF. This analysis aimed to better understand the impact of LPA1 antagonism on extracellular matrix (ECM)-neoepitope biomarkers and lung function through a post hoc analysis of the phase 2 study, along with an in vitro fibrogenesis model. METHODS Serum levels of nine ECM-neoepitope biomarkers were measured in patients with IPF. The association of biomarkers with baseline and change from baseline FVC and quantitative lung fibrosis as measured with high-resolution computed tomography, and differences between treatment arms using linear mixed models, were assessed. The Scar-in-a-Jar in vitro fibrogenesis model was used to further elucidate the antifibrotic mechanism of BMS-986020. RESULTS In 140 patients with IPF, baseline ECM-neoepitope biomarker levels did not predict FVC progression but was significantly correlated with baseline FVC and lung fibrosis measurements. Most serum ECM-neoepitope biomarker levels were significantly reduced following BMS-986020 treatment compared with placebo, and several of the reductions correlated with FVC and/or lung fibrosis improvement. In the Scar-in-a-Jar in vitro model, BMS-986020 potently inhibited LPA1-induced fibrogenesis. CONCLUSIONS BMS-986020 reduced serum ECM-neoepitope biomarkers, which were previously associated with IPF prognosis. In vitro, LPA promoted fibrogenesis, which was LPA1 dependent and inhibited by BMS-986020. Together these data elucidate a novel antifibrotic mechanism of action for pharmacological LPA1 blockade. Trial registration ClinicalTrials.gov identifier: NCT01766817; First posted: January 11, 2013; https://clinicaltrials.gov/ct2/show/NCT01766817 .
Collapse
Affiliation(s)
- Benjamin E Decato
- Research & Early Development, Bristol Myers Squibb, 3401 Princeton Pike, Princeton, NJ, 08648, USA
| | | | | | - Aryeh Fischer
- Research & Early Development, Bristol Myers Squibb, 3401 Princeton Pike, Princeton, NJ, 08648, USA
| | - Shuyan Du
- Research & Early Development, Bristol Myers Squibb, 3401 Princeton Pike, Princeton, NJ, 08648, USA
| | - Scott M Palmer
- Duke University Medical Center, 2085 Msrb2 2 Genome Ct., Durham, NC, 27710, USA
| | - Morten Karsdal
- Nordic Bioscience, Herlev Hovedgade 205-207, 2730 Herlev, Denmark
| | - Yi Luo
- Research & Early Development, Bristol Myers Squibb, 3401 Princeton Pike, Princeton, NJ, 08648, USA
| | - Anne Minnich
- Research & Early Development, Bristol Myers Squibb, 3401 Princeton Pike, Princeton, NJ, 08648, USA.
| |
Collapse
|
12
|
Khan MM, Poeckel D, Halavatyi A, Zukowska-Kasprzyk J, Stein F, Vappiani J, Sevin DC, Tischer C, Zinn N, Eley JD, Gudmann NS, Muley T, Winter H, Fisher AJ, Nanthakumar CB, Bergamini G, Pepperkok R. An integrated multiomic and quantitative label-free microscopy-based approach to study pro-fibrotic signalling in ex vivo human precision-cut lung slices. Eur Respir J 2021; 58:2000221. [PMID: 33361096 PMCID: PMC8318569 DOI: 10.1183/13993003.00221-2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 12/09/2020] [Indexed: 12/17/2022]
Abstract
Fibrosis can affect any organ, resulting in the loss of tissue architecture and function with often life-threatening consequences. Pathologically, fibrosis is characterised by the expansion of connective tissue due to excessive deposition of extracellular matrix (ECM) proteins, including the fibrillar forms of collagen. A significant limitation for discovering cures for fibrosis is the availability of suitable human models and techniques to quantify mature fibrillar collagen deposition as close as possible to human physiological conditions.Here we have extensively characterised an ex vivo cultured human lung tissue-derived, precision-cut lung slices (hPCLS) model using label-free second harmonic generation (SHG) light microscopy to quantify fibrillar collagen deposition and mass spectrometry-based techniques to obtain a proteomic and metabolomic fingerprint of hPCLS in ex vivo culture.We demonstrate that hPCLS are viable and metabolically active, with mesenchymal, epithelial, endothelial and immune cell types surviving for at least 2 weeks in ex vivo culture. Analysis of hPCLS-conditioned supernatants showed a strong induction of pulmonary fibrosis-related ECM proteins upon transforming growth factor-β1 (TGF-β1) stimulation. This upregulation of ECM proteins was not translated into an increased deposition of fibrillar collagen. In support of this observation, we revealed the presence of a pro-ECM degradation activity in our ex vivo cultures of hPCLS, inhibition of which by a metalloproteinase inhibitor resulted in increased collagen deposition in response to TGF-β1 stimulation.Together the data show that an integrated approach of measuring soluble pro-fibrotic markers alongside quantitative SHG-based analysis of fibrillar collagen is a valuable tool for studying pro-fibrotic signalling and testing anti-fibrotic agents.
Collapse
Affiliation(s)
- Muzamil Majid Khan
- European Molecular Biology Laboratory, Heidelberg, Germany
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Daniel Poeckel
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | - Aliaksandr Halavatyi
- European Molecular Biology Laboratory, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | | | - Frank Stein
- European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Daniel C Sevin
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | | | - Nico Zinn
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
| | | | | | - Thomas Muley
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Biobank Thoraxklinik, University Hospital Heidelberg, Heidelberg, Germany
| | - Hauke Winter
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Biobank Thoraxklinik, University Hospital Heidelberg, Heidelberg, Germany
| | - Andrew J Fisher
- Newcastle University Translational and Clinical Research Institute and Institute of Transplantation, Newcastle upon Tyne Hospitals, Newcastle upon Tyne, UK
| | | | - Giovanna Bergamini
- Discovery Biology, Cellzome GmbH, GSK, Heidelberg, Germany
- G. Bergamini and R. Pepperkok contributed equally to this article as lead authors and supervised the work
| | - Rainer Pepperkok
- European Molecular Biology Laboratory, Heidelberg, Germany
- Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- G. Bergamini and R. Pepperkok contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
13
|
Development of an Experimental Ex Vivo Wound Model to Evaluate Antimicrobial Efficacy of Topical Formulations. Int J Mol Sci 2021; 22:ijms22095045. [PMID: 34068733 PMCID: PMC8126222 DOI: 10.3390/ijms22095045] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/25/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Wound infections are considered a major cause for wound-associated morbidity. There is a high demand for alternative, robust, and affordable methods that can provide relatable and reproducible results when testing topical treatments, both in research and in the pharmaceutical industry. Here we present an ex vivo wound infection model using porcine skin and a burn wounding method, allowing for the efficacy evaluation of topical antimicrobial formulations. Utilizing this model, we demonstrate the potential of topical treatments after infecting the wounds with clinically significant bacteria, P. aeruginosa and S. aureus. We show that the method is compatible with several analytical tools used to analyze infection and antimicrobial effects. Both bacterial strains successfully infected the wound surface, as well as deeper regions of the tissue. Quantification of viable bacteria on the wound surface and in the tissue, longitudinal measurements of bioluminescence, fluorescence microscopy, and scanning electron microscopy were used to confirm the effects of antibacterial treatments. Furthermore, we show that biofilms are formed on the wound surface, indicating that the demonstrated method mirrors typical in vivo infections.
Collapse
|
14
|
Juhl P, Bondesen S, Hawkins CL, Karsdal MA, Bay-Jensen AC, Davies MJ, Siebuhr AS. Dermal fibroblasts have different extracellular matrix profiles induced by TGF-β, PDGF and IL-6 in a model for skin fibrosis. Sci Rep 2020; 10:17300. [PMID: 33057073 PMCID: PMC7560847 DOI: 10.1038/s41598-020-74179-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 09/28/2020] [Indexed: 12/11/2022] Open
Abstract
Different stimulants might induce different extracellular matrix profiles. It is essential to gain an understanding and quantification of these changes to allow for focused anti-fibrotic drug development. This study investigated the expression of extracellular matrix by dermal fibroblast mimicking fibrotic skin diseases as SSc using clinically validated biomarkers. Primary healthy human dermal fibroblasts were grown in media containing FICOLL. The cells were stimulated with PDGF-AB, TGF-β1, or IL-6. Anti-fibrotic compounds (iALK-5, Nintedanib) were added together with growth factors. Biomarkers of collagen formation and degradation together with fibronectin were evaluated by ELISAs in the collected supernatant. Immunohistochemical staining was performed to visualize fibroblasts and proteins, while selected gene expression levels were examined through qPCR. TGF-β and PDGF, and to a lesser extent IL-6, increased the metabolic activity of the fibroblasts. TGF-β primarily increased type I collagen and fibronectin protein and gene expression together with αSMA. PDGF stimulation resulted in increased type III and VI collagen formation and gene expression. IL-6 decreased fibronectin levels. iALK5 could inhibit TGF-β induced fibrosis while nintedanib could halt fibrosis induced by TGF-β or PDGF. Tocilizumab could not inhibit fibrosis induced in this model. The extent and nature of fibrosis are dependent on the stimulant. The model has potential as a pre-clinical model as the fibroblasts fibrotic phenotype could be reversed by an ALK5 inhibitor and Nintedanib.
Collapse
Affiliation(s)
- Pernille Juhl
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Biomarkers and Research, Nordic Bioscience, Herlev hovedgade 207, 2730, Herlev, Denmark.
| | - Sandie Bondesen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Biomarkers and Research, Nordic Bioscience, Herlev hovedgade 207, 2730, Herlev, Denmark
| | - Clare Louise Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Asser Karsdal
- Biomarkers and Research, Nordic Bioscience, Herlev hovedgade 207, 2730, Herlev, Denmark
| | | | | | - Anne Sofie Siebuhr
- Biomarkers and Research, Nordic Bioscience, Herlev hovedgade 207, 2730, Herlev, Denmark
| |
Collapse
|
15
|
Rønnow SR, Dabbagh RQ, Genovese F, Nanthakumar CB, Barrett VJ, Good RB, Brockbank S, Cruwys S, Jessen H, Sorensen GL, Karsdal MA, Leeming DJ, Sand JMB. Prolonged Scar-in-a-Jar: an in vitro screening tool for anti-fibrotic therapies using biomarkers of extracellular matrix synthesis. Respir Res 2020; 21:108. [PMID: 32381012 PMCID: PMC7203825 DOI: 10.1186/s12931-020-01369-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 04/22/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a rapidly progressing disease with challenging management. To find novel effective therapies, better preclinical models are needed for the screening of anti-fibrotic compounds. Activated fibroblasts drive fibrogenesis and are the main cells responsible for the accumulation of extracellular matrix (ECM). Here, a prolonged Scar-in-a-Jar assay was combined with clinically validated biochemical markers of ECM synthesis to evaluate ECM synthesis over time. To validate the model as a drug screening tool for novel anti-fibrotic compounds, two approved compounds for IPF, nintedanib and pirfenidone, and a compound in development, omipalisib, were tested. METHODS Primary human lung fibroblasts from healthy donors were cultured for 12 days in the presence of ficoll and were stimulated with TGF-β1 with or without treatment with an ALK5/TGF-β1 receptor kinase inhibitor (ALK5i), nintedanib, pirfenidone or the mTOR/PI3K inhibitor omipalisib (GSK2126458). Biomarkers of ECM synthesis were evaluated over time in cell supernatants using ELISAs to assess type I, III, IV, V and VI collagen formation (PRO-C1, PRO-C3, PRO-C4, PRO-C5, PRO-C6), fibronectin (FBN-C) deposition and α-smooth muscle actin (α-SMA) expression. RESULTS TGF-β1 induced synthesis of PRO-C1, PRO-C6 and FBN-C as compared with unstimulated fibroblasts at all timepoints, while PRO-C3 and α-SMA levels were not elevated until day 8. Elevated biomarkers were reduced by suppressing TGF-β1 signalling with ALK5i. Nintedanib and omipalisib were able to reduce all biomarkers induced by TGF-β1 in a concentration dependent manner, while pirfenidone had no effect on α-SMA. CONCLUSIONS TGF-β1 stimulated synthesis of type I, III and VI collagen, fibronectin and α-SMA but not type IV or V collagen. Synthesis was increased over time, although temporal profiles differed, and was modulated pharmacologically by ALK5i, nintedanib, pirfenidone and omipalisib. This prolonged 12-day Scar-in-a-Jar assay utilising biochemical markers of ECM synthesis provides a useful screening tool for novel anti-fibrotic compounds.
Collapse
Affiliation(s)
- Sarah Rank Rønnow
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rand Qais Dabbagh
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | - Federica Genovese
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | | | - Vikki J Barrett
- Department of Fibrosis DPU, Respiratory TA, GlaxoSmithKline, Stevenage, UK
| | - Robert B Good
- Department of Fibrosis DPU, Respiratory TA, GlaxoSmithKline, Stevenage, UK
| | - Sarah Brockbank
- Innovative Medicines Unit, Grünenthal Innovation, Aachen, Germany
- Present Address: Medicines Discovery Catapult, Alderley Edge, Cheshire, UK
| | - Simon Cruwys
- Innovative Medicines Unit, Grünenthal Innovation, Aachen, Germany
- Present Address: TherapeutAix AG, Aachen, Germany
| | - Henrik Jessen
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | - Grith Lykke Sorensen
- Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Morten Asser Karsdal
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | - Diana Julie Leeming
- Nordic Bioscience A/S, Herlev, Herlev Hovedgade 205-207, DK-2730, Herlev, Denmark
| | | |
Collapse
|
16
|
Cirrhotic Human Liver Extracellular Matrix 3D Scaffolds Promote Smad-Dependent TGF-β1 Epithelial Mesenchymal Transition. Cells 2019; 9:cells9010083. [PMID: 31905709 PMCID: PMC7017194 DOI: 10.3390/cells9010083] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/12/2022] Open
Abstract
An altered liver microenvironment characterized by a dysregulated extracellular matrix (ECM) supports the development and progression of hepatocellular carcinoma (HCC). The development of experimental platforms able to reproduce these physio-pathological conditions is essential in order to identify and validate new therapeutic targets for HCC. The aim of this work was to validate a new in vitro model based on engineering three-dimensional (3D) healthy and cirrhotic human liver scaffolds with HCC cells recreating the micro-environmental features favoring HCC. Healthy and cirrhotic human livers ECM scaffolds were developed using a high shear stress oscillation-decellularization procedure. The scaffolds bio-physical/bio-chemical properties were analyzed by qualitative and quantitative approaches. Cirrhotic 3D scaffolds were characterized by biomechanical properties and microarchitecture typical of the native cirrhotic tissue. Proteomic analysis was employed on decellularized 3D scaffolds and showed specific enriched proteins in cirrhotic ECM in comparison to healthy ECM proteins. Cell repopulation of cirrhotic scaffolds highlighted a unique up-regulation in genes related to epithelial to mesenchymal transition (EMT) and TGFβ signaling. This was also supported by the presence and release of higher concentration of endogenous TGFβ1 in cirrhotic scaffolds in comparison to healthy scaffolds. Fibronectin secretion was significantly upregulated in cells grown in cirrhotic scaffolds in comparison to cells engrafted in healthy scaffolds. TGFβ1 induced the phosphorylation of canonical proteins Smad2/3, which was ECM scaffold-dependent. Important, TGFβ1-induced phosphorylation of Smad2/3 was significantly reduced and ECM scaffold-independent when pre/simultaneously treated with the TGFβ-R1 kinase inhibitor Galunisertib. In conclusion, the inherent features of cirrhotic human liver ECM micro-environment were dissected and characterized for the first time as key pro-carcinogenic components in HCC development.
Collapse
|
17
|
Guan S, Zhang K, Li J. Recent Advances in Extracellular Matrix for Engineering Stem Cell Responses. Curr Med Chem 2019; 26:6321-6338. [DOI: 10.2174/0929867326666190704121309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/02/2018] [Accepted: 01/25/2019] [Indexed: 02/06/2023]
Abstract
Stem cell transplantation is an advanced medical technology, which brings hope for the
treatment of some difficult diseases in the clinic. Attributed to its self-renewal and differential
ability, stem cell research has been pushed to the forefront of regenerative medicine and has become
a hot topic in tissue engineering. The surrounding extracellular matrix has physical functions
and important biological significance in regulating the life activities of cells, which may play crucial
roles for in situ inducing specific differentiation of stem cells. In this review, we discuss the
stem cells and their engineering application, and highlight the control of the fate of stem cells, we
offer our perspectives on the various challenges and opportunities facing the use of the components
of extracellular matrix for stem cell attachment, growth, proliferation, migration and differentiation.
Collapse
Affiliation(s)
- Shuaimeng Guan
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou 450000, China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
18
|
Tian HY, Chen JY, Lin J, Liang QR, Lei Y, Li X, Wu Y, Yang LY, Lin XH, Liu AL, Chen YZ. Sepsis progression monitoring via human serum fibronectin detection based on sandwich-type electrochemical immunosensor. Anal Chim Acta 2019; 1100:225-231. [PMID: 31987145 DOI: 10.1016/j.aca.2019.11.068] [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: 06/29/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
Sepsis has always been a severe clinical problem in critical care medicine due to its rather high mortality and poor prognosis. The current study reported for the first time a practical immunosensor for fibronectin (FN) detection in human serum by electrochemistry. A simple but robust sandwich-type strategy was employed without any complex design or material modifications, which exhibited a linear calibration plot over the 15.625-500 ng/mL concentration range and a detection limit of 15 ng/mL. The results showed that the proposed strategy displayed an excellent selectivity against other non-target substances in human serum, a higher accuracy and a better stability when compared with the traditional enzyme-linked immunosorbent assay (ELISA) in detecting the same or mixed serum from 21 healthy subjects. Furthermore, the proposed electrochemical immunosensor successfully monitored the level of serum FN at various time points in five septic patients during the treatment. These findings demonstrate that the proposed strategy is highly sensitive and accurate in monitoring sepsis progress and has significant clinical improvements over the ELISA methodology, signifying a great potential of a commercial kit.
Collapse
Affiliation(s)
- Hui-Yun Tian
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Jin-Yuan Chen
- The Centralab, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China; Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Jia Lin
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Qi-Rui Liang
- Fujian Key Laboratory of Medical Bioinformatics, Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350122, China
| | - Yun Lei
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Xin Li
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Yong Wu
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Liang-Yong Yang
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| | - Xin-Hua Lin
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Ai-Lin Liu
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China.
| | - Yuan-Zhong Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
19
|
Nissen NI, Karsdal M, Willumsen N. Post-translational modifications of vimentin reflect different pathological processes associated with non-small cell lung cancer and chronic obstructive pulmonary disease. Oncotarget 2019; 10:6829-6841. [PMID: 31827725 PMCID: PMC6887574 DOI: 10.18632/oncotarget.27332] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/06/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Vimentin has shown to be highly implicated in cancer initiation and progression. Vimentin is often a target of post-translational modifications (PTMs) which can be disease specific, thus targeting these specific modifications can be of high biomarker potential. In this study we set out to evaluate the biological relevance and serum biomarker potential of citrullinated vimentin (VICM) and non-citrullinated vimentin (VIM) in non-small cell lung cancer (NSCLC) and chronic obstructive pulmonary disease (COPD). METHODS A competitive ELISA targeting VIM was developed and quantified in serum from patients with NSCLC and COPD. VIM was compared with levels of VICM in the same indications. RESULTS VIM was significantly increased in NSCLC (n = 100) compared to healthy controls (n = 67) in two independent cohorts (p = 0.0003 and p < 0.0001). Furthermore, VIM was highly increased in late stages of NSCLC (p = 0.001), however VIM was not increased in COPD patients (n = 10). Contrarily, serum levels of VICM was not increased in late stages of NSCLC, but highly elevated in patients with COPD (p < 0.0001). CONCLUSIONS These findings suggest a biomarker potential of VIM in NSCLC. Our findings also indicate that PTMs of vimentin are highly relevant and that targeting these modifications can have differential biomarker potential.
Collapse
Affiliation(s)
- Neel Ingemann Nissen
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark.,Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | - Morten Karsdal
- Nordic Bioscience, Biomarkers and Research, DK-2730 Herlev, Denmark
| | | |
Collapse
|
20
|
Koszegi S, Molnar A, Lenart L, Hodrea J, Balogh DB, Lakat T, Szkibinszkij E, Hosszu A, Sparding N, Genovese F, Wagner L, Vannay A, Szabo AJ, Fekete A. RAAS inhibitors directly reduce diabetes-induced renal fibrosis via growth factor inhibition. J Physiol 2018; 597:193-209. [PMID: 30324679 PMCID: PMC6312411 DOI: 10.1113/jp277002] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Increased activation of the renin-angiotensin-aldosterone system (RAAS) and elevated growth factor production are of crucial importance in the development of renal fibrosis leading to diabetic kidney disease. The aim of this study was to provide evidence for the antifibrotic potential of RAAS inhibitor (RAASi) treatment and to explore the exact mechanism of this protective effect. We found that RAASi ameliorate diabetes-induced renal interstitial fibrosis and decrease profibrotic growth factor production. RAASi prevents fibrosis by acting directly on proximal tubular cells, and inhibits hyperglycaemia-induced growth factor production and thereby fibroblast activation. These results suggest a novel therapeutic indication and potential of RAASi in the treatment of renal fibrosis. ABSTRACT In diabetic kidney disease (DKD) increased activation of renin-angiotensin-aldosterone system (RAAS) contributes to renal fibrosis. Although RAAS inhibitors (RAASi) are the gold standard therapy in DKD, the mechanism of their antifibrotic effect is not yet clarified. Here we tested the antifibrotic and renoprotective action of RAASi in a rat model of streptozotocin-induced DKD. In vitro studies on proximal tubular cells and renal fibroblasts were also performed to further clarify the signal transduction pathways that are directly altered by hyperglycaemia. After 5 weeks of diabetes, male Wistar rats were treated for two more weeks per os with the RAASi ramipril, losartan, spironolactone or eplerenone. Proximal tubular cells were cultured in normal or high glucose (HG) medium and treated with RAASi. Platelet-derived growth factor (PDGF) or connective tissue growth factor (CTGF/CCN2)-induced renal fibroblasts were also treated with various RAASi. In diabetic rats, reduced renal function and interstitial fibrosis were ameliorated and elevated renal profibrotic factors (TGFβ1, PDGF, CTGF/CCN2, MMP2, TIMP1) and alpha-smooth muscle actin (αSMA) levels were decreased by RAASi. HG increased growth factor production of HK-2 cells, which in turn induced activation and αSMA production of fibroblasts. RAASi decreased tubular PDGF and CTGF expression and reduced production of extracellular matrix (ECM) components in fibroblasts. In proximal tubular cells, hyperglycaemia-induced growth factor production increased renal fibroblast transformation, contributing to the development of fibrosis. RAASi, even in non-antihypertensive doses, decreased the production of profibrotic factors and directly prevented fibroblast activation. All these findings suggest a novel therapeutic role for RAASi in the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Sandor Koszegi
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Agnes Molnar
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,1st Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Lilla Lenart
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Judit Hodrea
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Dora Bianka Balogh
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,1st Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Tamas Lakat
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Edgar Szkibinszkij
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Adam Hosszu
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,1st Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - Nadja Sparding
- Nordic Bioscience, Biomarkers & Research, Herlev, Denmark.,Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | | | - Laszlo Wagner
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Adam Vannay
- MTA-SE Paediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Attila J Szabo
- 1st Department of Paediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Paediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Andrea Fekete
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.,1st Department of Paediatrics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
21
|
Sorrell JM, Somoza RA, Caplan AI. Human mesenchymal stem cells induced to differentiate as chondrocytes follow a biphasic pattern of extracellular matrix production. J Orthop Res 2018; 36:1757-1766. [PMID: 29194731 PMCID: PMC5976510 DOI: 10.1002/jor.23820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/27/2017] [Indexed: 02/04/2023]
Abstract
Regenerative medicine and tissue engineering studies are actively developing novel means to repair adult articular cartilage defects using biological approaches. One such approach is the harnessing of adult human therapeutic cells such as those referred to as mesenchymal stem cells. Upon exposure to chondrogenic signals, these cells differentiate and initiate the production of a complex and voluminous cartilaginous matrix that is crucial to both the structure and function of cartilage. Furthermore, this complexity requires the time-sensitive activation of a large number of genes to produce the components of this matrix. The current study analyzed the kinetics of matrix production in an aggregate culture model where adult human mesenchymal stem cells were induced to differentiate as chondrocytes. The results indicate the existence of a biphasic mode of differentiation and maturation during which matrix genes and molecules are differentially activated and secreted. These results have important implications for developing novel approaches for the creation of tissue engineered articular cartilage. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1757-1766, 2018.
Collapse
Affiliation(s)
- J. Michael Sorrell
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Rodrigo A. Somoza
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| | - Arnold I. Caplan
- Department of Biology, Skeletal Research Center; Case Western Reserve University; Cleveland Ohio 44106
| |
Collapse
|
22
|
Kaessmeyer S, Sehl J, Khiao In M, Merle R, Richardson K, Plendl J. Subcellular Interactions during Vascular Morphogenesis in 3D Cocultures between Endothelial Cells and Fibroblasts. Int J Mol Sci 2017; 18:ijms18122590. [PMID: 29194374 PMCID: PMC5751193 DOI: 10.3390/ijms18122590] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/23/2017] [Accepted: 11/28/2017] [Indexed: 12/28/2022] Open
Abstract
Background: Increasing the complexity of in vitro systems to mimic three-dimensional tissues and the cellular interactions within them will increase the reliability of data that were previously collected with in vitro systems. In vivo vascularization is based on complex and clearly defined cell–matrix and cell–cell interactions, where the extracellular matrix (ECM) seems to play a very important role. The aim of this study was to monitor and visualize the subcellular and molecular interactions between endothelial cells (ECs), fibroblasts, and their surrounding microenvironment during vascular morphogenesis in a three-dimensional coculture model. Methods: Quantitative and qualitative analyses during the generation of a coculture tissue construct consisting of endothelial cells and fibroblasts were done using transmission electron microscopy and immunohistochemistry. Results: Dynamic interactions were found in cocultures between ECs, between fibroblasts (FBs), between ECs and FBs, and between the cells and the ECM. Microvesicles were involved in intercellular information transfer. FBs took an active and physical part in the angiogenesis process. The ECM deposited by the cells triggered endothelial angiogenic activity. Capillary-like tubular structures developed and matured. Moreover, some ECM assembled into a basement membrane (BM) having three different layers equivalent to those seen in vivo. Finally, the three-dimensional in vitro construct mirrored the topography of histological tissue sections. Conclusion: Our results visualize the importance of the physical contact between all cellular and acellular components of the cocultures.
Collapse
Affiliation(s)
- Sabine Kaessmeyer
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Julia Sehl
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Maneenooch Khiao In
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| | - Roswitha Merle
- Department of Veterinary Medicine, Institute of Veterinary Epidemiology and Biostatistics, Freie Universitaet Berlin, Koenigsweg 67, 14163 Berlin, Germany.
| | - Ken Richardson
- College of Veterinary Medicine, School of Veterinary and Life Sciences, Murdoch University, Murdoch, WA 6150, Australia.
| | - Johanna Plendl
- Department of Veterinary Medicine, Institute of Veterinary Anatomy, Freie Universitaet Berlin, Koserstraße 20, 14195 Berlin, Germany.
| |
Collapse
|