1
|
Wang M, Zhang J, Qiao C, Yan S, Wu G. Comparative analysis of human and mouse transcriptomes during skin wound healing. Front Cell Dev Biol 2024; 12:1486493. [PMID: 39534398 PMCID: PMC11554618 DOI: 10.3389/fcell.2024.1486493] [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: 08/28/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Skin wound healing is a complex process which involves multiple molecular events and the underlying mechanism is not fully understood. We presented a comparative transcriptomic analysis of skin wound healing in humans and mice to identify shared molecular mechanisms across species. We analyzed transcriptomes from three distinct stages of the healing process and constructed protein-protein interaction networks to elucidate commonalities in the healing process. A substantial number of differentially expressed genes (DEGs) were identified in human transcriptomes, particularly upregulated genes before and after wound injury, and enriched in processes related to extracellular matrix organization and leukocyte migration. Similarly, the mouse transcriptome revealed thousands of DEGs, with shared biological processes and enriched KEGG pathways, highlighting a conserved molecular signature in skin wound healing. A total of 21 common DEGs were found across human comparisons, and 591 in mouse comparisons, with four genes (KRT2, MARCKSL1, MMP1, and TNC) consistently differentially expressed in both species, suggesting critical roles in mammalian skin wound healing. The expression trends of these genes were consistent, indicating their potential as therapeutic targets. The molecular network analysis identified five subnetworks associated with collagen synthesis, immunity, cell-cell adhesion, and extracellular matrix, with hub genes such as COL4A1, TLR7, TJP3, MMP13, and HIF1A exhibited significant expression changes before and after wound injury in humans and mice. In conclusion, our study provided a detailed molecular network for understanding the healing process in humans and mice, revealing conserved mechanisms that could help the development of targeted therapies across species.
Collapse
Affiliation(s)
- Maochun Wang
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | - Guoping Wu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
2
|
Sun C, Bai M, Jia Y, Tian X, Guo Y, Xu X, Guo Z. mRNA sequencing reveals the distinct gene expression and biological functions in cardiac fibroblasts regulated by recombinant fibroblast growth factor 2. PeerJ 2023; 11:e15736. [PMID: 37483983 PMCID: PMC10362857 DOI: 10.7717/peerj.15736] [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: 02/09/2023] [Accepted: 06/20/2023] [Indexed: 07/25/2023] Open
Abstract
After myocardial injury, cardiac fibroblasts (CFs) differentiate into myofibroblasts, which express and secrete extracellular matrix (ECM) components for myocardial repair, but also promote myocardial fibrosis. Recombinant fibroblast growth factor 2 (FGF2) protein drug with low molecular weight can promote cell survival and angiogenesis, and it was found that FGF2 could inhibit the activation of CFs, suggesting FGF2 has great potential in myocardial repair. However, the regulatory role of FGF2 on CFs has not been fully elucidated. Here, we found that recombinant FGF2 significantly suppressed the expression of alpha smooth muscle actin (α-SMA) in CFs. Through RNA sequencing, we analyzed mRNA expression in CFs and the differently expressed genes regulated by FGF2, including 430 up-regulated genes and 391 down-regulated genes. Gene ontology analysis revealed that the differentially expressed genes were strongly enriched in multiple biological functions, including ECM organization, cell adhesion, actin filament organization and axon guidance. The results of gene set enrichment analysis (GSEA) show that ECM organization and actin filament organization are down-regulated, while axon guidance is up-regulated. Further cellular experiments indicate that the regulatory functions of FGF2 are consistent with the findings of the gene enrichment analysis. This study provides valuable insights into the potential therapeutic role of FGF2 in treating cardiac fibrosis and establishes a foundation for further research to uncover the underlying mechanisms of CFs gene expression regulated by FGF2.
Collapse
Affiliation(s)
- Changye Sun
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Mengru Bai
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yangyang Jia
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiangqin Tian
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yonglong Guo
- Department of Cardiology, The First Affiliated Hospital, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xinhui Xu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhikun Guo
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan, China
| |
Collapse
|
3
|
Fibroblast growth factor-2 bound to specific dermal fibroblast-derived extracellular vesicles is protected from degradation. Sci Rep 2022; 12:22131. [PMID: 36550142 PMCID: PMC9780220 DOI: 10.1038/s41598-022-26217-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Fibroblast growth factor-2 (FGF2) has multiple roles in cutaneous wound healing but its natural low stability prevents the development of its use in skin repair therapies. Here we show that FGF2 binds the outer surface of dermal fibroblast (DF)-derived extracellular vesicles (EVs) and this association protects FGF2 from fast degradation. EVs isolated from DF cultured in the presence of FGF2 harbor FGF2 on their surface and FGF2 can bind purified EVs in absence of cells. Remarkably, FGF2 binding to EVs is restricted to a specific subpopulation of EVs, which do not express CD63 and CD81 markers. Treatment of DF with FGF2-EVs activated ERK and STAT signaling pathways and increased cell proliferation and migration. Local injection of FGF2-EVs improved wound healing in mice. We further demonstrated that binding to EVs protects FGF2 from both thermal and proteolytic degradation, thus maintaining FGF2 function. This suggests that EVs protect soluble factors from degradation and increase their stability and half-life. These results reveal a novel aspect of EV function and suggest EVs as a potential tool for delivering FGF2 in skin healing therapies.
Collapse
|
4
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
5
|
Ground M, Waqanivavalagi S, Park YE, Callon K, Walker R, Milsom P, Cornish J. Fibroblast growth factor 2 inhibits myofibroblastic activation of valvular interstitial cells. PLoS One 2022; 17:e0270227. [PMID: 35714127 PMCID: PMC9205485 DOI: 10.1371/journal.pone.0270227] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/06/2022] [Indexed: 12/30/2022] Open
Abstract
Heart valve disease is a growing problem worldwide. Though very common in older adults, the mechanisms behind the development of the disease aren't well understood, and at present the only therapeutic option is valve replacement. Valvular interstitial cells (VICs) may hold the answer. These cells can undergo pathological differentiation into contractile myofibroblasts or osteoblasts, leading to thickening and calcification of the valve tissue. Our study aimed to characterise the effect of fibroblast growth factor 2 (FGF-2) on the differentiation potential of VICs. We isolated VICs from diseased human valves and treated these cells with FGF-2 and TGF-β to elucidate effect of these growth factors on several myofibroblastic outcomes, in particular immunocytochemistry and gene expression. We used TGF-β as a positive control for myofibroblastic differentiation. We found that FGF-2 promotes a 'quiescent-type' morphology and inhibits the formation of α-smooth muscle actin positive myofibroblasts. FGF-2 reduced the calcification potential of VICs, with a marked reduction in the number of calcific nodules. FGF-2 interrupted the 'canonical' TGF-β signalling pathway, reducing the nuclear translocation of the SMAD2/3 complex. The panel of genes assayed revealed that FGF-2 promoted a quiescent-type pattern of gene expression, with significant downregulations in typical myofibroblast markers α smooth muscle actin, extracellular matrix proteins, and scleraxis. We did not see evidence of osteoblast differentiation: neither matrix-type calcification nor changes in osteoblast associated gene expression were observed. Our findings show that FGF-2 can reverse the myofibroblastic phenotype of VICs isolated from diseased valves and inhibit the calcification potential of these cells.
Collapse
Affiliation(s)
- Marcus Ground
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Steve Waqanivavalagi
- Green Lane Cardiothoracic Surgery Unit, Auckland City Hospital, Auckland District Health Board, Grafton, New Zealand
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Young-Eun Park
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Karen Callon
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| | - Robert Walker
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Paget Milsom
- Green Lane Cardiothoracic Surgery Unit, Auckland City Hospital, Auckland District Health Board, Grafton, New Zealand
| | - Jillian Cornish
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Grafton, New Zealand
| |
Collapse
|
6
|
Dolivo DM. Anti-fibrotic effects of pharmacologic FGF-2: a review of recent literature. J Mol Med (Berl) 2022; 100:847-860. [PMID: 35484303 DOI: 10.1007/s00109-022-02194-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/09/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023]
Abstract
Fibrosis is a process of pathological tissue repair that replaces damaged, formerly functional tissue with a non-functional, collagen-rich scar. Complications of fibrotic pathologies, which can arise in numerous organs and from numerous conditions, result in nearly half of deaths in the developed world. Despite this, therapies that target fibrosis at its mechanistic roots are still notably lacking. The ubiquity of the occurrence of fibrosis in myriad organs emphasizes the fact that there are shared mechanisms underlying fibrotic conditions, which may serve as common therapeutic targets for multiple fibrotic diseases of varied organs. Thus, study of the basic science of fibrosis and of anti-fibrotic modalities is critical to therapeutic development and may have potential to translate across organs and disease states. Fibroblast growth factor 2 (FGF-2) is a broadly studied member of the fibroblast growth factors, a family of multipotent cytokines implicated in diverse cellular and tissue processes, which has previously been recognized for its anti-fibrotic potential. However, the mechanisms underlying this potential are not fully understood, nor is the potential for its use to ameliorate fibrosis in diverse pathologies and tissues. Presented here is a review of recent literature that sheds further light on these questions, with the hopes of inspiring further research into the mechanisms underlying the anti-fibrotic activities of FGF-2, as well as the disease conditions for which pharmacologic FGF-2 might be a useful option in the future.
Collapse
|
7
|
Deshpande N, Wilkins MR, Khachigian LM. RNA sequencing identifies genes reliant upon Ser26 in early growth response-1 in vascular endothelial cells exposed to fibroblast growth factor-2. Vascul Pharmacol 2022; 143:106952. [PMID: 35041980 DOI: 10.1016/j.vph.2022.106952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/08/2022] [Indexed: 11/25/2022]
Abstract
Early growth response-1 (Egr-1) is an inducible master regulatory transcription factor that orchestrates gene expression in vascular endothelial cells. We recently determined that Ser26 in Egr-1 undergoes phosphorylation and plays a critical functional role in a range of pro-angiogenic processes. To better understand the effect of Ser26 on Egr-1-dependent gene expression, in this study, we performed RNA-seq and bioinformatics analysis on human microvascular endothelial cells bearing a germline mutation (M) in Ser26 to Ala (M26 cells) exposed to the mitogen and chemoattractant fibroblast growth factor-2 (FGF2) as compared with wildtype (WT) cells. In WT cells, FGF2 increased the expression of numerous growth factors and hormones cytokines, signaling molecules and transcriptional regulators. Comparison of FGF2-inducible WT and M26 cells enabled identification of differentially expressed genes, including genes reliant or not reliant upon Ser26. For example, Ser26 in Egr-1 was required for FGF2 inducible LIF expression but not for FGF2 inducible IL11. Ser26 was also required for FGF2 inducible NKX2-8 and RIPK2 expression but not for FGF2 inducible CREB5 or ALPK2 expression. Conversely, FGF2 inhibited genes such as TIE1, GPR146 and EPHB3, and Ser26 was required for FGF2's effect on TIE1 and GPR146 but not for EPHB3. Enrichment analysis also identified a range of gene ontologies upregulated and downregulated by FGF2. These findings demonstrate the importance of Ser26 in Egr-1 in programs of endothelial gene expression modulated by FGF2.
Collapse
Affiliation(s)
- Nandan Deshpande
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Marc R Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Levon M Khachigian
- Vascular Biology and Translational Research, School of Medical Sciences & UNSW Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
8
|
Paskal W, Kopka M, Stachura A, Paskal AM, Pietruski P, Pełka K, Woessner AE, Quinn KP, Galus R, Wejman J, Włodarski P. Single Dose of N-Acetylcysteine in Local Anesthesia Increases Expression of HIF1α, MAPK1, TGFβ1 and Growth Factors in Rat Wound Healing. Int J Mol Sci 2021; 22:8659. [PMID: 34445365 PMCID: PMC8395485 DOI: 10.3390/ijms22168659] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 01/13/2023] Open
Abstract
In this study, we aimed to investigate the influence of N-acetylcysteine (NAC) on the gene expression profile, neoangiogenesis, neutrophils and macrophages in a rat model of incisional wounds. Before creating wounds on the backs of 24 Sprague-Dawley rats, intradermal injections were made. Lidocaine-epinephrin solutions were supplemented with 0.015%, 0.03% or 0.045% solutions of NAC, or nothing (control group). Scars were harvested on the 3rd, 7th, 14th and 60th day post-surgery. We performed immunohistochemical staining in order to visualize macrophages (anti-CD68), neutrophils (anti-MPO) and newly formed blood vessels (anti-CD31). Additionally, RT-qPCR was used to measure the relative expression of 88 genes involved in the wound healing process. On the 14th day, the number of cells stained with anti-CD68 and anti-CD31 antibodies was significantly larger in the tissues treated with 0.03% NAC compared with the control. Among the selected genes, 52 were upregulated and six were downregulated at different time points. Interestingly, NAC exerted a significant effect on the expression of 45 genes 60 days after its administration. In summation, a 0.03% NAC addition to the pre-incisional anesthetic solution improves neovasculature and increases the macrophages' concentration at the wound site on the 14th day, as well as altering the expression of numerous genes that are responsible for the regenerative processes.
Collapse
Affiliation(s)
- Wiktor Paskal
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
| | - Michał Kopka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
| | - Albert Stachura
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
- Doctoral School, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adriana M. Paskal
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
| | - Piotr Pietruski
- Centre of Postgraduate Medical Education, Department of Replantation and Reconstructive Surgery, Gruca Teaching Hospital, 05-400 Otwock, Poland;
| | - Kacper Pełka
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
| | - Alan E. Woessner
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (A.E.W.); (K.P.Q.)
| | - Kyle P. Quinn
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA; (A.E.W.); (K.P.Q.)
| | - Ryszard Galus
- Department of Histology and Embryology, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Jarosław Wejman
- Department of Pathology, Centre of Postgraduate Medical Education, 00-416 Warsaw, Poland;
| | - Paweł Włodarski
- Department of Methodology, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (A.S.); (A.M.P.); (K.P.); (P.W.)
| |
Collapse
|
9
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
10
|
Deciphering the role of cartilage protein 1 in human dermal fibroblasts: a transcriptomic approach. Funct Integr Genomics 2021; 21:503-511. [PMID: 34269961 DOI: 10.1007/s10142-021-00792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 10/20/2022]
Abstract
Cartilage acidic protein 1A (hCRTAC1-A) is an extracellular matrix protein (ECM) of human hard and soft tissue that is associated with matrix disorders. The central role of fibroblasts in tissue integrity and ECM health made primary human dermal fibroblasts (NHDF) the model for the present study, which aimed to provide new insight into the molecular function of hCRTAC1-A. Specifically, we explored the differential expression patterns of specific genes associated with the presence of hCRTAC1-A by RNA-seq and RT-qPCR analysis. Functional enrichment analysis demonstrated, for the very first time, that hCRTAC1-A is involved in extracellular matrix organization and development, through its regulatory effect on asporin, decorin, and complement activity, in cell proliferation, regeneration, wound healing, and collagen degradation. This work provides a better understanding of putative hCRTAC1-A actions in human fibroblasts and a fundamental insight into its function in tissue biology.
Collapse
|
11
|
Functional Properties of a Purified Reconstituted Bilayer Matrix Design Support Natural Wound Healing Activities. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2021; 9:e3596. [PMID: 34036030 PMCID: PMC8140771 DOI: 10.1097/gox.0000000000003596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/09/2021] [Indexed: 12/26/2022]
Abstract
Biomaterial engineering has produced numerous matrices for use in tissue repair, utilizing various materials and processing methods, which can impact the ability of the products to encourage wound healing. Recently, we observed favorable clinical outcomes, using a novel purified reconstituted bilayer matrix (PRBM; Geistlich Derma-Gide) to treat chronic diabetic foot ulcers.
Collapse
|
12
|
Wu B, Tang X, Zhou Z, Ke H, Tang S, Ke R. RNA sequencing analysis of FGF2-responsive transcriptome in skin fibroblasts. PeerJ 2021; 9:e10671. [PMID: 33520460 PMCID: PMC7812929 DOI: 10.7717/peerj.10671] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/08/2020] [Indexed: 01/01/2023] Open
Abstract
Background Fibroblast growth factor 2 (FGF2) is a highly pleiotropic cytokine with antifibrotic activity in wound healing. During the process of wound healing and fibrosis, fibroblasts are the key players. Although accumulating evidence has suggested the antagonistic effects of FGF2 in the activation process of fibroblasts, the mechanisms by which FGF2 hinders the fibroblast activation remains incompletely understood. This study aimed to identify the key genes and their regulatory networks in skin fibroblasts treated with FGF2. Methods RNA-seq was performed to identify the differentially expressed mRNA (DEGs) and lncRNA between FGF2-treated fibroblasts and control. DEGs were analyzed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Furthermore, the networks between mRNAs and lncRNAs were constructed by Pearson correlation analysis and the networkanalyst website. Finally, hub genes were validated by real time-PCR. Results Between FGF2-treated fibroblasts and control fibroblasts, a total of 1475 DEGs was obtained. These DEGs were mainly enriched in functions such as the ECM organization, cell adhesion, and cell migration. They were mainly involved in ECM-receptor interaction, PI3K-Akt signaling, and the Hippo pathway. The hub DEGs included COL3A1, COL4A1, LOX, PDGFA, TGFBI, and ITGA10. Subsequent real-time PCR, as well as bioinformatics analysis, consistently demonstrated that the expression of ITGA10 was significantly upregulated while the other five DEGs (COL3A1, COL4A1, LOX, PDGFA, TGFBI) were downregulated in FGF2-treated fibroblasts. Meanwhile, 213 differentially expressed lncRNAs were identified and three key lncRNAs (HOXA-AS2, H19, and SNHG8) were highlighted in FGF2-treated fibroblasts. Conclusion The current study comprehensively analyzed the FGF2-responsive transcriptional profile and provided candidate mechanisms that may account for FGF2-mediated wound healing.
Collapse
Affiliation(s)
- Baojin Wu
- Department of Plastic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Xinjie Tang
- Department of Plastic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhaoping Zhou
- Department of Plastic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Honglin Ke
- Department of Emergency, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Shao Tang
- Department of Statistics, Florida State University, Tallahassee, FL, USA
| | - Ronghu Ke
- Department of Plastic Surgery, Huashan Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
13
|
Alibardi L. Appendage regeneration in anamniotes utilizes genes active during larval-metamorphic stages that have been lost or altered in amniotes: The case for studying lizard tail regeneration. J Morphol 2020; 281:1358-1381. [PMID: 32865265 DOI: 10.1002/jmor.21251] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/20/2020] [Accepted: 07/25/2020] [Indexed: 12/17/2022]
Abstract
This review elaborates the idea that organ regeneration derives from specific evolutionary histories of vertebrates. Regenerative ability depends on genomic regulation of genes specific to the life-cycles that have differentially evolved in anamniotes and amniotes. In aquatic environments, where fish and amphibians live, one or multiple metamorphic transitions occur before the adult stage is reached. Each transition involves the destruction and remodeling of larval organs that are replaced with adult organs. After organ injury or loss in adult anamniotes, regeneration uses similar genes and developmental process than those operating during larval growth and metamorphosis. Therefore, the broad presence of regenerative capability across anamniotes is possible because generating new organs is included in their life history at metamorphic stages. Soft hyaluronate-rich regenerative blastemas grow in submersed or in hydrated environments, that is, essential conditions for regeneration, like during development. In adult anamniotes, the ability to regenerate different organs decreases in comparison to larval stages and becomes limited during aging. Comparisons of genes activated during metamorphosis and regeneration in anamniotes identify key genes unique to these processes, and include thyroid, wnt and non-coding RNAs developmental pathways. In the terrestrial environment, some genes or developmental pathways for metamorphic transitions were lost during amniote evolution, determining loss of regeneration. Among amniotes, the formation of soft and hydrated blastemas only occurs in lizards, a morphogenetic process that evolved favoring their survival through tail autotomy, leading to a massive although imperfect regeneration of the tail. Deciphering genes activity during lizard tail regeneration would address future attempts to recreate in other amniotes regenerative blastemas that grow into variably completed organs.
Collapse
|
14
|
Wang P, Wang H, Ma K, Wang S, Yang C, Mu N, Yang F, Feng H, Chen T. Novel cytokine-loaded PCL-PEG scaffold composites for spinal cord injury repair. RSC Adv 2020; 10:6306-6314. [PMID: 35495987 PMCID: PMC9049693 DOI: 10.1039/c9ra10385f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 01/20/2020] [Indexed: 12/14/2022] Open
Abstract
Severe spinal cord injury (SCI) always leads to permanent sensory and motor dysfunction. However, the therapeutic effects of current treatment methods, including high dose methylprednisolone, surgical interventions and rehabilitative care, are far from satisfactory. In recent years, cellular, molecular, tissue engineering and rehabilitative training have shown promising results in animal models. Poly-ε-caprolacton (PCL) - based hydrogel composite system has been considered as a promising strategy to direct the axon growth and mimic the properties of natural extracellular matrix. In this study, we found the addition of the fibroblast growth factor 2 (FGF2) and epidermal growth factor (EGF) to the hydrogel induces the production of axon growth-supportive substrates. The addition of the glial-derived neurotrophic factor (GDNF) to the hydrogel further induces axon directional growth. This "five-in-one" composite scaffold, referred to as PCL/PEG/FGF2/EGF/GDNF, improved the locomotor function in rats 8 weeks after spinal cord injury (SCI) after implantation in transected spinal cord. Furthermore, histological assessment indicated that the designed composite scaffold guided the neuronal regeneration and promoted the production of axon growth-supportive substrates, providing a favorable biological microenvironment. Our novel composite scaffold provides a promising therapeutic method for SCI.
Collapse
Affiliation(s)
- Pangbo Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Hufei Wang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Kang Ma
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Shi Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Chuanyan Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Ning Mu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Fei Yang
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics & Chemistry, Institute of Chemistry Chinese Academy of Sciences Beijing 100190 China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| | - Tunan Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University) Chongqing 400038 China
| |
Collapse
|
15
|
Angiotensin-II-Evoked Ca 2+ Entry in Murine Cardiac Fibroblasts Does Not Depend on TRPC Channels. Cells 2020; 9:cells9020322. [PMID: 32013125 PMCID: PMC7072683 DOI: 10.3390/cells9020322] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/23/2020] [Accepted: 01/25/2020] [Indexed: 02/06/2023] Open
Abstract
TRPC proteins form cation conducting channels regulated by different stimuli and are regulators of the cellular calcium homeostasis. TRPC are expressed in cardiac cells including cardiac fibroblasts (CFs) and have been implicated in the development of pathological cardiac remodeling including fibrosis. Using Ca2+ imaging and several compound TRPC knockout mouse lines we analyzed the involvement of TRPC proteins for the angiotensin II (AngII)-induced changes in Ca2+ homeostasis in CFs isolated from adult mice. Using qPCR we detected transcripts of all Trpc genes in CFs; Trpc1, Trpc3 and Trpc4 being the most abundant ones. We show that the AngII-induced Ca2+ entry but also Ca2+ release from intracellular stores are critically dependent on the density of CFs in culture and are inversely correlated with the expression of the myofibroblast marker α-smooth muscle actin. Our Ca2+ measurements depict that the AngII- and thrombin-induced Ca2+ transients, and the AngII-induced Ca2+ entry and Ca2+ release are not affected in CFs isolated from mice lacking all seven TRPC proteins (TRPC-hepta KO) compared to control cells. However, pre-incubation with GSK7975A (10 µM), which sufficiently inhibits CRAC channels in other cells, abolished AngII-induced Ca2+ entry. Consequently, we conclude the dispensability of the TRPC channels for the acute neurohumoral Ca2+ signaling evoked by AngII in isolated CFs and suggest the contribution of members of the Orai channel family as molecular constituents responsible for this pathophysiologically important Ca2+ entry pathway.
Collapse
|
16
|
Anderson MA, O'Shea TM, Burda JE, Ao Y, Barlatey SL, Bernstein AM, Kim JH, James ND, Rogers A, Kato B, Wollenberg AL, Kawaguchi R, Coppola G, Wang C, Deming TJ, He Z, Courtine G, Sofroniew MV. Required growth facilitators propel axon regeneration across complete spinal cord injury. Nature 2018; 561:396-400. [PMID: 30158698 PMCID: PMC6151128 DOI: 10.1038/s41586-018-0467-6] [Citation(s) in RCA: 349] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/13/2018] [Indexed: 12/17/2022]
Abstract
Transected axons fail to regrow across anatomically complete spinal cord injuries (SCI) in adults. Diverse molecules can partially facilitate or attenuate axon growth during development or after injury1-3, but efficient reversal of this regrowth failure remains elusive4. Here we show that three factors that are essential for axon growth during development but are attenuated or lacking in adults-(i) neuron intrinsic growth capacity2,5-9, (ii) growth-supportive substrate10,11 and (iii) chemoattraction12,13-are all individually required and, in combination, are sufficient to stimulate robust axon regrowth across anatomically complete SCI lesions in adult rodents. We reactivated the growth capacity of mature descending propriospinal neurons with osteopontin, insulin-like growth factor 1 and ciliary-derived neurotrophic factor before SCI14,15; induced growth-supportive substrates with fibroblast growth factor 2 and epidermal growth factor; and chemoattracted propriospinal axons with glial-derived neurotrophic factor16,17 delivered via spatially and temporally controlled release from biomaterial depots18,19, placed sequentially after SCI. We show in both mice and rats that providing these three mechanisms in combination, but not individually, stimulated robust propriospinal axon regrowth through astrocyte scar borders and across lesion cores of non-neural tissue that was over 100-fold greater than controls. Stimulated, supported and chemoattracted propriospinal axons regrew a full spinal segment beyond lesion centres, passed well into spared neural tissue, formed terminal-like contacts exhibiting synaptic markers and conveyed a significant return of electrophysiological conduction capacity across lesions. Thus, overcoming the failure of axon regrowth across anatomically complete SCI lesions after maturity required the combined sequential reinstatement of several developmentally essential mechanisms that facilitate axon growth. These findings identify a mechanism-based biological repair strategy for complete SCI lesions that could be suitable to use with rehabilitation models designed to augment the functional recovery of remodelling circuits.
Collapse
Affiliation(s)
- Mark A Anderson
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Timothy M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Joshua E Burda
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sabry L Barlatey
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Alexander M Bernstein
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jae H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Nicholas D James
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Alexandra Rogers
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Brian Kato
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexander L Wollenberg
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Giovanni Coppola
- Departments of Psychiatry and Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chen Wang
- F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Timothy J Deming
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Department of Neurology, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gregoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Ji J, He D, Feng Y, He Y, Xue F, Xie L. JDINAC: joint density-based non-parametric differential interaction network analysis and classification using high-dimensional sparse omics data. Bioinformatics 2018; 33:3080-3087. [PMID: 28582486 DOI: 10.1093/bioinformatics/btx360] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 06/01/2017] [Indexed: 12/26/2022] Open
Abstract
Motivation A complex disease is usually driven by a number of genes interwoven into networks, rather than a single gene product. Network comparison or differential network analysis has become an important means of revealing the underlying mechanism of pathogenesis and identifying clinical biomarkers for disease classification. Most studies, however, are limited to network correlations that mainly capture the linear relationship among genes, or rely on the assumption of a parametric probability distribution of gene measurements. They are restrictive in real application. Results We propose a new Joint density based non-parametric Differential Interaction Network Analysis and Classification (JDINAC) method to identify differential interaction patterns of network activation between two groups. At the same time, JDINAC uses the network biomarkers to build a classification model. The novelty of JDINAC lies in its potential to capture non-linear relations between molecular interactions using high-dimensional sparse data as well as to adjust confounding factors, without the need of the assumption of a parametric probability distribution of gene measurements. Simulation studies demonstrate that JDINAC provides more accurate differential network estimation and lower classification error than that achieved by other state-of-the-art methods. We apply JDINAC to a Breast Invasive Carcinoma dataset, which includes 114 patients who have both tumor and matched normal samples. The hub genes and differential interaction patterns identified were consistent with existing experimental studies. Furthermore, JDINAC discriminated the tumor and normal sample with high accuracy by virtue of the identified biomarkers. JDINAC provides a general framework for feature selection and classification using high-dimensional sparse omics data. Availability and implementation R scripts available at https://github.com/jijiadong/JDINAC. Contact lxie@iscb.org. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jiadong Ji
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Di He
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA
| | - Yang Feng
- Department of Statistics, Columbia University, New York, NY 10027, USA
| | - Yong He
- Department of Mathematical Statistics, School of Statistics, Shandong University of Finance and Economics, Jinan 250014, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Shandong University, Jinan 250012, China
| | - Lei Xie
- Ph.D. Program in Computer Science, The Graduate Center, The City University of New York, New York, NY 10016, USA.,Department of Computer Science, Hunter College, The City University of New York, NY 10065, USA
| |
Collapse
|
18
|
Dolivo D, Hernandez S, Dominko T. Cellular lifespan and senescence: a complex balance between multiple cellular pathways. Bioessays 2017; 38 Suppl 1:S33-44. [PMID: 27417120 DOI: 10.1002/bies.201670906] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 11/09/2022]
Abstract
The study of cellular senescence and proliferative lifespan is becoming increasingly important because of the promises of autologous cell therapy, the need for model systems for tissue disease and the implication of senescent cell phenotypes in organismal disease states such as sarcopenia, diabetes and various cancers, among others. Here, we explain the concepts of proliferative cellular lifespan and cellular senescence, and we present factors that have been shown to mediate cellular lifespan positively or negatively. We review much recent literature and present potential molecular mechanisms by which lifespan mediation occurs, drawing from the fields of telomere biology, metabolism, NAD(+) and sirtuin biology, growth factor signaling and oxygen and antioxidants. We conclude that cellular lifespan and senescence are complex concepts that are governed by multiple independent and interdependent pathways, and that greater understanding of these pathways, their interactions and their convergence upon specific cellular phenotypes may lead to viable therapies for tissue regeneration and treatment of age-related pathologies, which are caused by or exacerbated by senescent cells in vivo.
Collapse
Affiliation(s)
- David Dolivo
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Sarah Hernandez
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Tanja Dominko
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| |
Collapse
|
19
|
Dolivo DM, Larson SA, Dominko T. Fibroblast Growth Factor 2 as an Antifibrotic: Antagonism of Myofibroblast Differentiation and Suppression of Pro-Fibrotic Gene Expression. Cytokine Growth Factor Rev 2017; 38:49-58. [PMID: 28967471 DOI: 10.1016/j.cytogfr.2017.09.003] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 09/22/2017] [Indexed: 02/08/2023]
Abstract
Fibrosis is a pathological condition that is characterized by the replacement of dead or damaged tissue with a nonfunctional, mechanically aberrant scar, and fibrotic pathologies account for nearly half of all deaths worldwide. The causes of fibrosis differ somewhat from tissue to tissue and pathology to pathology, but in general some of the cellular and molecular mechanisms remain constant regardless of the specific pathology in question. One of the common mechanisms underlying fibroses is the paradigm of the activated fibroblast, termed the "myofibroblast," a differentiated mesenchymal cell with demonstrated contractile activity and a high rate of collagen deposition. Fibroblast growth factor 2 (FGF2), one of the members of the mammalian fibroblast growth factor family, is a cytokine with demonstrated antifibrotic activity in non-human animal, human, and in vitro models. FGF2 is highly pleiotropic and its receptors are present on many different cell types throughout the body, lending a great deal of variety to the potential mechanisms of FGF2 effects on fibrosis. However, recent reports demonstrate that a substantial contribution to the antifibrotic effects of FGF2 comes from the inhibitory effects of FGF2 on connective tissue fibroblasts, activated myofibroblasts, and myofibroblast progenitors. FGF2 demonstrates effects antagonistic towards fibroblast activation and towards mesenchymal transition of potential myofibroblast-forming cells, as well as promotes a gene expression paradigm more reminiscent of regenerative healing, such as that which occurs in the fetal wound healing response, than fibrotic resolution. With a better understanding of the mechanisms by which FGF2 alters the wound healing cascade and results in a shift away from scar formation and towards functional tissue regeneration, we may be able to further address the critical need of therapy for varied fibrotic pathologies across myriad tissue types.
Collapse
Affiliation(s)
- David M Dolivo
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States
| | - Sara A Larson
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States
| | - Tanja Dominko
- Worcester Polytechnic Institute, Department of Biology and Biotechnology,100 Institute Road, Worcester, MA, 01609, United States.
| |
Collapse
|
20
|
Veber M, Dolivo D, Rolle M, Dominko T. Pro-myogenic and low-oxygen culture increases expression of contractile smooth muscle markers in human fibroblasts. J Tissue Eng Regen Med 2017; 12:572-582. [PMID: 28513058 DOI: 10.1002/term.2473] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/25/2017] [Accepted: 05/09/2017] [Indexed: 01/23/2023]
Abstract
Smooth muscle cells (SMCs) are essential for tissue engineering strategies to fabricate organs such as blood vessels, the oesophagus and bladder, and to create disease models of these systems. In order for such therapies and models to be feasible, SMCs must be sourced effectively to enable production of large numbers of functional cells. In vitro, SMCs divide slowly and demonstrate short proliferative lifespans compared with other types of cells, including stem cells and fibroblasts, limiting the number of cells that can be derived from expansion in culture of a primary isolation. As such, it would be beneficial to better understand the factors underlying induction and maintenance of SMC phenotypes, in order to produce new sources of SMCs for tissue engineering and disease modelling. Here we report the ability of human dermal fibroblasts to display patterns of gene expression resembling contractile SMCs when cultured under conditions that are known to promote a contractile phenotype in SMCs, including culture on collagen IV, low-serum culture, TGF-β1 treatment and hypoxia. These factors drive expression of the myogenic transcription factor myocardin, as well as expression of several of its gene targets that are known contributors to contractile phenotype in SMCs, including smooth muscle alpha actin, calponin, and myosin heavy chain. Our results suggest that culture conditions associated with culture of SMCs may be sufficient to induce myogenic gene expression patterns and potential myogenic function in non-muscle cells.
Collapse
Affiliation(s)
| | - David Dolivo
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Marsha Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, USA
| | - Tanja Dominko
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, USA.,Center for Biomedical Sciences and Engineering, University of Nova Gorica, Vipava, Slovenia
| |
Collapse
|
21
|
Dolivo DM, Larson SA, Dominko T. FGF2-mediated attenuation of myofibroblast activation is modulated by distinct MAPK signaling pathways in human dermal fibroblasts. J Dermatol Sci 2017; 88:339-348. [PMID: 28899582 DOI: 10.1016/j.jdermsci.2017.08.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/18/2017] [Accepted: 08/25/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Previous human and animal studies have demonstrated the ability of exogenously administered basic fibroblast growth factor (FGF2) to act as an antifibrotic agent in the skin. Though the activity of FGF2 as an anti-scarring agent is well-established for fibrotic skin wounds, the mechanisms by which FGF2 exerts these actions are not entirely understood. Canonical FGF2 signaling proceeds in part via FGFR/MAPK pathways in human dermal fibroblasts, and FGF2 has been described to prevent or reverse the fibroblast-to-myofibroblast transition, which is driven by TGFβ signaling and understood to be an important step in the formation of a fibrotic scar in vivo. Thus, we set out to investigate the antagonistic effects of FGF2 on TGFβ signaling as well as the broader effects of MAPK inhibition on the TGFβ-mediated induction of myofibroblast gene expression. OBJECTIVE To better understand the effects of FGF2 signaling pathways on myofibroblastic gene expression and cell phenotypes. METHODS Human dermal fibroblasts were cultured in vitro in the presence of FGF2, TGFβ, and/or MAPK inhibitors, and the effects of these agents were investigated by molecular biology techniques including qRT-PCR, immunofluorescence, Western blot, and flow cytometry. RESULTS FGF2 inhibited TGFβ-mediated fibroblast activation, resulting in more rapidly proliferating, spindle-shaped cells, compared to the more slowly proliferating, flatter TGFβ-treated cells. Treatment with FGF2 also attenuated TGFβ-mediated increase in expression of myofibroblast markers smooth muscle α-actin, calponin, transgelin, connective tissue growth factor, ED-A fibronectin, and collagen I. FGF2-mediated antagonism of the TGFβ-mediated fibroblast-to-myofibroblast transition was reversed by small molecule inhibition of ERK or JNK, and it was potentiated by inhibition of p38. MAPK inhibition was demonstrated to have qualitatively similar effects even in the absence of exogenous FGF2, and small molecule inhibition of p38 MAPK was sufficient to attenuate TGFβ-mediated fibroblast activation. CONCLUSIONS Inhibition of select MAPK signaling pathways can reverse or potentiate anti-fibrotic FGF2 effects on human dermal fibroblasts, as well as exert their effects independently of exogenous FGF2 supplementation.
Collapse
Affiliation(s)
- David M Dolivo
- Worcester Polytechnic Institute, Department of Biology and Biotechnology, 100 Institute Road, Worcester, MA, 01609, United States
| | - Sara A Larson
- Worcester Polytechnic Institute, Department of Biology and Biotechnology, 100 Institute Road, Worcester, MA, 01609, United States
| | - Tanja Dominko
- Worcester Polytechnic Institute, Department of Biology and Biotechnology, 100 Institute Road, Worcester, MA, 01609, United States.
| |
Collapse
|
22
|
Chung H, Multhaupt HAB, Oh ES, Couchman JR. Minireview: Syndecans and their crucial roles during tissue regeneration. FEBS Lett 2016; 590:2408-17. [PMID: 27383370 DOI: 10.1002/1873-3468.12280] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/30/2022]
Abstract
Syndecans are transmembrane heparan sulfate proteoglycans, with roles in development, tumorigenesis and inflammation, and growing evidence for involvement in tissue regeneration. This is a fast developing field with the prospect of utilizing tissue engineering and biomaterials in novel therapies. Syndecan receptors are not only ubiquitous in mammalian tissues, regulating cell adhesion, migration, proliferation, and differentiation through independent signaling but also working alongside other receptors. Their importance is highlighted by an ability to interact with a diverse array of ligands, including extracellular matrix glycoproteins, growth factors, morphogens, and cytokines that are important regulators of regeneration. We also discuss the potential for syndecans to regulate stem cell properties, and suggest that understanding these proteoglycans is relevant to exploiting cell, tissue, and materials technologies.
Collapse
Affiliation(s)
- Heesung Chung
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - Hinke A B Multhaupt
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| | - Eok-Soo Oh
- Department of Life Sciences and the Research Center for Cellular Homeostasis, Ewha Womans University, Seoul, Korea
| | - John R Couchman
- Department of Biomedical Sciences and Biotech Research & Innovation Center, University of Copenhagen, Denmark
| |
Collapse
|
23
|
Lichtenberger BM, Mastrogiannaki M, Watt FM. Epidermal β-catenin activation remodels the dermis via paracrine signalling to distinct fibroblast lineages. Nat Commun 2016; 7:10537. [PMID: 26837596 PMCID: PMC4742837 DOI: 10.1038/ncomms10537] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/23/2015] [Indexed: 01/06/2023] Open
Abstract
Sustained epidermal Wnt/β-catenin signalling expands the stem cell compartment and induces ectopic hair follicles (EFs). This is accompanied by extensive fibroblast proliferation and extracellular matrix (ECM) remodelling in the underlying dermis. Here we show that epidermal Hedgehog (Hh) and Transforming growth factor-beta (TGF-β) signalling mediate the dermal changes. Pharmacological inhibition or genetic deletion of these pathways prevents β-catenin-induced dermal reprogramming and EF formation. Epidermal Shh stimulates proliferation of the papillary fibroblast lineage, whereas TGF-β2 controls proliferation, differentiation and ECM production by reticular fibroblasts. Hh inhibitors do not affect TGF-β target gene expression in reticular fibroblasts, and TGF-β inhibition does not prevent Hh target gene induction in papillary fibroblasts. However, when Hh signalling is inhibited the reticular dermis does not respond to epidermal β-catenin activation. We conclude that the dermal response to epidermal Wnt/β-catenin signalling depends on distinct fibroblast lineages responding to different paracrine signals. The molecular mechanisms regulating skin dermal changes are unclear. Here, the authors show that deletion of Hedgehog (Hh) in the upper dermis alters the response to epidermal Wnt signalling, which, together with changes in extracellular matrix production, influences distinct fibroblast lineages differently.
Collapse
Affiliation(s)
- Beate M Lichtenberger
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,Wellcome Trust Centre for Stem Cell Research, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK
| | - Maria Mastrogiannaki
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| |
Collapse
|
24
|
Sideek MA, Teia A, Kopecki Z, Cowin AJ, Gibson MA. Co-localization of LTBP-2 with FGF-2 in fibrotic human keloid and hypertrophic scar. J Mol Histol 2015; 47:35-45. [PMID: 26644005 DOI: 10.1007/s10735-015-9645-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/25/2015] [Indexed: 02/06/2023]
Abstract
We have recently shown that Latent transforming growth factor-beta-1 binding protein-2 (LTBP-2) has a single high-affinity binding site for fibroblast growth factor-2 (FGF-2) and that LTBP-2 blocks FGF-2 induced cell proliferation. Both proteins showed strong co-localisation within keloid skin from a single patient. In the current study, using confocal microscopy, we have investigated the distribution of the two proteins in normal and fibrotic skin samples including normal scar tissue, hypertrophic scars and keloids from multiple patients. Consistently, little staining for either protein was detected in normal adult skin and normal scar samples but extensive co-localisation of the two proteins was observed in multiple examples of hypertrophic scars and keloids. LTBP-2 and FGF-2 were co-localised to fine fibrous elements within the extracellular matrix identified as elastic fibres by immunostaining with anti-fibrillin-1 and anti-elastin antibodies. Furthermore, qPCR analysis of RNA samples from multiple patients confirmed dramatically increased expression of LTBP-2 and FGF-2, similar TGF-beta 1, in hypertrophic scar compared to normal skin and scar tissue. Overall the results suggest that elevated LTBP-2 may bind and sequester FGF-2 on elastic fibres in fibrotic tissues and modulate FGF-2's influence on the repair and healing processes.
Collapse
Affiliation(s)
- Mohamed A Sideek
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia.,Department of Biomedical Sciences, Kulliyyah of Allied Health Sciences, International Islamic University Malaysia, 25200, Kuantan, Pahang, Malaysia
| | - Abdulrahman Teia
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Zlatko Kopecki
- Regenerative Medicine, Mawson Institute, University of South Australia, Adelaide, SA, 5095, Australia
| | - Allison J Cowin
- Regenerative Medicine, Mawson Institute, University of South Australia, Adelaide, SA, 5095, Australia
| | - Mark A Gibson
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia.
| |
Collapse
|
25
|
Karbiener M, Glantschnig C, Pisani DF, Laurencikiene J, Dahlman I, Herzig S, Amri EZ, Scheideler M. Mesoderm-specific transcript (MEST) is a negative regulator of human adipocyte differentiation. Int J Obes (Lond) 2015; 39:1733-41. [PMID: 26119994 PMCID: PMC4625608 DOI: 10.1038/ijo.2015.121] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 06/09/2015] [Accepted: 06/22/2015] [Indexed: 11/16/2022]
Abstract
BACKGROUND A growing body of evidence suggests that many downstream pathologies of obesity are amplified or even initiated by molecular changes within the white adipose tissue (WAT). Such changes are the result of an excessive expansion of individual white adipocytes and could potentially be ameliorated via an increase in de novo adipocyte recruitment (adipogenesis). Mesoderm-specific transcript (MEST) is a protein with a putative yet unidentified enzymatic function and has previously been shown to correlate with adiposity and adipocyte size in mouse. OBJECTIVES This study analysed WAT samples and employed a cell model of adipogenesis to characterise MEST expression and function in human. METHODS AND RESULTS MEST mRNA and protein levels increased during adipocyte differentiation of human multipotent adipose-derived stem cells. Further, obese individuals displayed significantly higher MEST levels in WAT compared with normal-weight subjects, and MEST was significantly correlated with adipocyte volume. In striking contrast to previous mouse studies, knockdown of MEST enhanced human adipocyte differentiation, most likely via a significant promotion of peroxisome proliferator-activated receptor signalling, glycolysis and fatty acid biosynthesis pathways at early stages. Correspondingly, overexpression of MEST impaired adipogenesis. We further found that silencing of MEST fully substitutes for the phosphodiesterase inhibitor 3-isobutyl-1-methylxanthine (IBMX) as an inducer of adipogenesis. Accordingly, phosphorylation of the pro-adipogenic transcription factors cyclic AMP responsive element binding protein (CREB) and activating transcription factor 1 (ATF1) were highly increased on MEST knockdown. CONCLUSIONS Although we found a similar association between MEST and adiposity as previously described for mouse, our functional analyses suggest that MEST acts as an inhibitor of human adipogenesis, contrary to previous murine studies. We have further established a novel link between MEST and CREB/ATF1 that could be of general relevance in regulation of metabolism, in particular obesity-associated diseases.
Collapse
Affiliation(s)
- M Karbiener
- Department of Phoniatrics, ENT University Hospital, Medical University Graz, Graz, Austria
| | - C Glantschnig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - D F Pisani
- Université Nice Sophia Antipolis, iBV, UMR, Nice, France
- CNRS, iBV, UMR, Nice, France
- Inserm, iBV, Nice, France
| | - J Laurencikiene
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - I Dahlman
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - S Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - E-Z Amri
- Université Nice Sophia Antipolis, iBV, UMR, Nice, France
- CNRS, iBV, UMR, Nice, France
- Inserm, iBV, Nice, France
| | - M Scheideler
- Institute for Diabetes and Cancer (IDC), Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
26
|
Grella A, Kole D, Holmes W, Dominko T. FGF2 Overrides TGFβ1-Driven Integrin ITGA11 Expression in Human Dermal Fibroblasts. J Cell Biochem 2015; 117:1000-8. [PMID: 26403263 DOI: 10.1002/jcb.25386] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 09/23/2015] [Indexed: 12/28/2022]
Abstract
Deposition of collagen-based extracellular matrix by fibroblasts during wound healing leads to scar formation--a typical outcome of the healing process in soft tissue wounds. The process can, however, be skewed in favor of tissue regeneration by manipulation of wound environment. Low oxygen conditions and supplementation with FGF2 provide extracellular cues that drive wound fibroblasts towards a pro-regenerative phenotype. Under these conditions, fibroblasts dramatically alter expression of many genes among which the most significantly deregulated are extracellular matrix and adhesion molecules. Here we investigate the mechanism of a collagen I binding integrin α11 (ITGA11) deregulation in response to low oxygen-mediated FGF2 effects in dermal fibroblasts. Using RT-PCR, qRT-PCR, Western blotting, and immunocytochemistry, we describe significant down-regulation of ITGA11. Decrease in ITGA11 is associated with its loss from focal adhesions. We show that loss of ITGA11 requires FGF2 induced ERK1/2 activity and in the presence of FGF2, ITGA11 expression cannot be rescued by TGFβ1, a potent activator of ITGA11. Our results indicate that FGF2 may be redirecting fibroblasts towards an anti-fibrotic phenotype by overriding TGFβ1 mediated ITGA11 expression.
Collapse
Affiliation(s)
- Alexandra Grella
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts, 01609
| | - Denis Kole
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts, 01609
| | - William Holmes
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts, 01609
| | - Tanja Dominko
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, 100 Institute Road, Worcester, Massachusetts, 01609.,Center for Biomedical Sciences and Engineering, University of Nova Gorica, Nova Gorica 5000, Slovenia
| |
Collapse
|
27
|
Gene Signature of Human Oral Mucosa Fibroblasts: Comparison with Dermal Fibroblasts and Induced Pluripotent Stem Cells. BIOMED RESEARCH INTERNATIONAL 2015; 2015:121575. [PMID: 26339586 PMCID: PMC4538314 DOI: 10.1155/2015/121575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 04/03/2015] [Accepted: 04/10/2015] [Indexed: 01/27/2023]
Abstract
Oral mucosa is a useful material for regeneration therapy with the advantages of its accessibility and versatility regardless of age and gender. However, little is known about the molecular characteristics of oral mucosa. Here we report the first comparative profiles of the gene signatures of human oral mucosa fibroblasts (hOFs), human dermal fibroblasts (hDFs), and hOF-derived induced pluripotent stem cells (hOF-iPSCs), linking these with biological roles by functional annotation and pathway analyses. As a common feature of fibroblasts, both hOFs and hDFs expressed glycolipid metabolism-related genes at higher levels compared with hOF-iPSCs. Distinct characteristics of hOFs compared with hDFs included a high expression of glycoprotein genes, involved in signaling, extracellular matrix, membrane, and receptor proteins, besides a low expression of HOX genes, the hDFs-markers. The results of the pathway analyses indicated that tissue-reconstructive, proliferative, and signaling pathways are active, whereas senescence-related genes in p53 pathway are inactive in hOFs. Furthermore, more than half of hOF-specific genes were similarly expressed to those of hOF-iPSC genes and might be controlled by WNT signaling. Our findings demonstrated that hOFs have unique cellular characteristics in specificity and plasticity. These data may provide useful insight into application of oral fibroblasts for direct reprograming.
Collapse
|
28
|
Kim MS, Yun JI, Gong SP, Ahn JY, Lim JM, Song YH, Park KH, Lee ST. Development of a chemically defined in vitro culture system to effectively stimulate the proliferation of adult human dermal fibroblasts. Exp Dermatol 2015; 24:543-5. [PMID: 25808127 DOI: 10.1111/exd.12695] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2015] [Indexed: 01/03/2023]
Abstract
Despite the fact that dermal fibroblasts are a practical model for research related to cell physiology and cell therapy, an in vitro culture system excluding serum, which complicates standardization and specificity and induces variability and unwanted effects, does not exist. We tried to establish a CDCS that supports effective proliferation of aHDFs. KDMEM supplemented with 5% (v/v) KSR, 12 ng/ml bFGF, 5 ng/ml EGF and 1 μg/ml hydrocortisone supported sufficient proliferation of aHDFs for 1 week. However, aHDF proliferation was decreased greatly after subculture. This problem could be overcome by culturing aHDFs in CDCM in culture plates coated with 10 μg/ml FN. Long-term culture of aHDFs was achieved using CDCM and FN-coated culture plates for 7 weeks. The optimized CDCS increased the proliferation of aHDFs significantly, without any increase in the senescence rate or alteration in morphology of aHDFs, despite long-term culture. In conclusion, we established a CDCS that improved proliferation of aHDFs while inhibiting cellular senescence. The CDCS will contribute to advances in various future research related to clinical skin regeneration.
Collapse
Affiliation(s)
- Min Seong Kim
- Department of Animal Life Science, Kangwon National University, Chuncheon, Korea
| | - Jung Im Yun
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Korea
| | - Seung Pyo Gong
- Department of Marine Biomaterials and Aquaculture, Pukyong National University, Busan, Korea
| | - Ji Yeon Ahn
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Korea
| | - Jeong Mook Lim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, Korea
| | - Young Han Song
- Department of Animal Life Science, Kangwon National University, Chuncheon, Korea.,Division of Animal Resource Science, Kangwon National University, Chuncheon, Korea
| | - Kyu Hyun Park
- Department of Animal Life Science, Kangwon National University, Chuncheon, Korea.,Division of Animal Resource Science, Kangwon National University, Chuncheon, Korea
| | - Seung Tae Lee
- Department of Animal Life Science, Kangwon National University, Chuncheon, Korea.,Division of Applied Animal Science, Kangwon National University, Chuncheon, Korea
| |
Collapse
|
29
|
Seguin L, Gozo M, Weis SM, Cheresh DA. Targeting the Achilles' heel of drug-resistant cancer stem cells. Cell Cycle 2014; 13:2017-8. [PMID: 24906000 PMCID: PMC4111689 DOI: 10.4161/cc.29460] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Affiliation(s)
- Laetitia Seguin
- Department of Pathology and Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| | - Maricel Gozo
- Department of Pathology and Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| | - Sara M Weis
- Department of Pathology and Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| | - David A Cheresh
- Department of Pathology and Moores Cancer Center; University of California, San Diego; La Jolla CA USA
| |
Collapse
|
30
|
Jez M, Ambady S, Kashpur O, Grella A, Malcuit C, Vilner L, Rozman P, Dominko T. Expression and differentiation between OCT4A and its Pseudogenes in human ESCs and differentiated adult somatic cells. PLoS One 2014; 9:e89546. [PMID: 24586860 PMCID: PMC3933561 DOI: 10.1371/journal.pone.0089546] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 01/22/2014] [Indexed: 12/02/2022] Open
Abstract
The POU5F1 gene codes for the OCT4 transcription factor, which is one of the key regulators of pluripotency. Its transcription, alternative splicing, and alternative translation leading to the synthesis of the active, nuclear localized OCT4A has been described in detail. Much less, however, is known about actively transcribed OCT4 pseudogenes, several of which display high homology to OCT4A and can be expressed and translated into proteins. Using RT-PCR followed by pseudogene-specific restriction digestion, cloning, and sequencing we discriminate between OCT4A and transcripts for pseudogenes 1, 3 and 4. We show that expression of OCT4 and its pseudogenes follows a developmentally-regulated pattern in differentiating hESCs, indicating a tight regulatory relationship between them. We further demonstrate that differentiated human cells from a variety of tissues express exclusively pseudogenes. Expression of OCT4A can, however be triggered in adult differentiated cells by oxygen and FGF2-dependent mechanisms.
Collapse
Affiliation(s)
- Mojca Jez
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Sakthikumar Ambady
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
| | - Olga Kashpur
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
| | - Alexandra Grella
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
| | - Christopher Malcuit
- Bioengineering Institute, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
- CellThera, Inc., Worcester, Massachusetts, United States of America
| | - Lucy Vilner
- CellThera, Inc., Worcester, Massachusetts, United States of America
| | - Primoz Rozman
- Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| | - Tanja Dominko
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
- Bioengineering Institute, Worcester Polytechnic Institute, Worcester, Massachusetts, United States of America
- CellThera, Inc., Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|