1
|
Jiang W, Pang X, Ha P, Li C, Chang GX, Zhang Y, Bossong LA, Ting K, Soo C, Zheng Z. Fibromodulin selectively accelerates myofibroblast apoptosis in cutaneous wounds by enhancing interleukin 1β signaling. Nat Commun 2025; 16:3499. [PMID: 40221432 PMCID: PMC11993684 DOI: 10.1038/s41467-025-58906-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Activated myofibroblasts deposit extracellular matrix material to facilitate rapid wound closure that can heal scarlessly during fetal development. However, adult myofibroblasts exhibit a relatively long life and persistent function, resulting in scarring. Thus, understanding how fetal and adult tissue regeneration differs may serve to identify factors that promote more optimal wound healing in adults with little or less scarring. We previously found that matricellular proteoglycan fibromodulin is one such factor promoting more optimal repair, but the underlying molecular and cellular mechanisms for these effects have not been fully elucidated. Here, we find that fibromodulin induces myofibroblast apoptosis after wound closure to reduce scarring in small and large animal models. Mechanistically, fibromodulin accelerates and prolongs the formation of the interleukin 1β-interleukin 1 receptor type 1-interleukin 1 receptor accessory protein ternary complex to increase the apoptosis of myofibroblasts and keloid- and hypertrophic scar-derived cells. As the persistence of myofibroblasts during tissue regeneration is a key cause of fibrosis in most organs, fibromodulin represents a promising, broad-spectrum anti-fibrotic therapeutic.
Collapse
Affiliation(s)
- Wenlu Jiang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Pin Ha
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Grace Xinlian Chang
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuxin Zhang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral, Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Lawrence A Bossong
- Department of Neuroscience, Princeton University, Princeton, NJ, 08540, USA
| | - Kang Ting
- American Dental Association Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, National Yang-Ming Chiao Tung University, Taipei, 30010, Taiwan.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
2
|
Zhao F, Wang L, Zhang Y, Tang S, Ji P, Xiang X, Pang X. MiR-494-3p regulates skin fibroblast activities by mediating fibromodulin production. J Cell Physiol 2024; 239:e31404. [PMID: 39129212 DOI: 10.1002/jcp.31404] [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: 02/07/2024] [Revised: 07/04/2024] [Accepted: 08/01/2024] [Indexed: 08/13/2024]
Abstract
Skin wound healing is a well-coordinated process in which various cells and factors participate, during which fibroblast exhibits a critical role by exerting its multiple activities, including proliferation, migration, invasion, and differentiation. Previous studies have identified that fibromodulin (FMOD) could enhance dermal wound healing by promoting skin fibroblast activities, but little is known about its upstream regulator. We occasionally found that FMOD expression was downregulated in skin fibroblast by transforming growth factor-β1 treatment. It was hypothesized that microRNAs (miRNA) in skin fibroblast could downregulate FMOD production and blocking them would increase FMOD expression, as well as promote skin wound healing. Here, by utilizing combined analysis of miRNA microarray from the Gene Expression Omnibus database and miRNA targets prediction, we successfully identified a miRNA, termed miR-494-3p, could regulate FMOD production in human skin fibroblast (BJ fibroblast). The functional analysis revealed that miR-494-3p mimics could inhibit BJ fibroblast migration and invasion but not proliferation and differentiation, while miR-494-3p inhibition markedly promotes migration, invasion, and differentiation of BJ fibroblast. Moreover, we established FMOD overexpression (OE) and knockout BJ fibroblast. We found that FMOD OE could rescue the inhibitory effects of miR-494-3p mimics on the migration and invasion of BJ fibroblast. In contrast, the miR-494-3p inhibitor transfection could not enhance migration, invasion, and differentiation of FMOD KO BJ fibroblast. Together, our results suggest that miR-494-3p may be a potential target for skin wound management via regulating FMOD production.
Collapse
Affiliation(s)
- Feng Zhao
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Linshu Wang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Yuxin Zhang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Siqi Tang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Ping Ji
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xuerong Xiang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoxiao Pang
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
3
|
Rehan IF, Elnagar A, Zigo F, Sayed-Ahmed A, Yamada S. Biomimetic strategies for the deputization of proteoglycan functions. Front Cell Dev Biol 2024; 12:1391769. [PMID: 39170918 PMCID: PMC11337302 DOI: 10.3389/fcell.2024.1391769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
Proteoglycans (PGs), which have glycosaminoglycan chains attached to their protein cores, are essential for maintaining the morphology and function of healthy body tissues. Extracellular PGs perform various functions, classified into the following four categories: i) the modulation of tissue mechanical properties; ii) the regulation and protection of the extracellular matrix; iii) protein sequestration; and iv) the regulation of cell signaling. The depletion of PGs may significantly impair tissue function, encompassing compromised mechanical characteristics and unregulated inflammatory responses. Since PGs play critical roles in the function of healthy tissues and their synthesis is complex, the development of PG mimetic molecules that recapitulate PG functions for tissue engineering and therapeutic applications has attracted the interest of researchers for more than 20 years. These approaches have ranged from semisynthetic graft copolymers to recombinant PG domains produced by cells that have undergone genetic modifications. This review discusses some essential extracellular PG functions and approaches to mimicking these functions.
Collapse
Affiliation(s)
- Ibrahim F. Rehan
- Department of Husbandry and Development of Animal Wealth, Faculty of Veterinary Medicine, Menoufia University, Shebin Alkom, Egypt
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - Asmaa Elnagar
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| | - František Zigo
- Department of Animal Nutrition and Husbandry, University of Veterinary Medicine and Pharmacy, Košice, Slovakia
| | - Ahmed Sayed-Ahmed
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Menoufia University, Shebin Alkom, Egypt
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, Nagoya, Aichi, Japan
| |
Collapse
|
4
|
Voza FA, Huerta CT, Le N, Shao H, Ribieras A, Ortiz Y, Atkinson C, Machuca T, Liu ZJ, Velazquez OC. Fibroblasts in Diabetic Foot Ulcers. Int J Mol Sci 2024; 25:2172. [PMID: 38396848 PMCID: PMC10889208 DOI: 10.3390/ijms25042172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/25/2024] Open
Abstract
Fibroblasts are stromal cells ubiquitously distributed in the body of nearly every organ tissue. These cells were previously considered to be "passive cells", solely responsible for ensuring the turnover of the extracellular matrix (ECM). However, their versatility, including their ability to switch phenotypes in response to tissue injury and dynamic activity in the maintenance of tissue specific homeostasis and integrity have been recently revealed by the innovation of technological tools such as genetically modified mouse models and single cell analysis. These highly plastic and heterogeneous cells equipped with multifaceted functions including the regulation of angiogenesis, inflammation as well as their innate stemness characteristics, play a central role in the delicately regulated process of wound healing. Fibroblast dysregulation underlies many chronic conditions, including cardiovascular diseases, cancer, inflammatory diseases, and diabetes mellitus (DM), which represent the current major causes of morbidity and mortality worldwide. Diabetic foot ulcer (DFU), one of the most severe complications of DM affects 40 to 60 million people. Chronic non-healing DFU wounds expose patients to substantial sequelae including infections, gangrene, amputation, and death. A complete understanding of the pathophysiology of DFU and targeting pathways involved in the dysregulation of fibroblasts are required for the development of innovative new therapeutic treatments, critically needed for these patients.
Collapse
Affiliation(s)
- Francesca A. Voza
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Carlos Theodore Huerta
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Nga Le
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Hongwei Shao
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antoine Ribieras
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Yulexi Ortiz
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carl Atkinson
- Department of Internal Medicine, Division of Pulmonary Critical Care & Sleep Medicine, University of Florida, Gainesville, FL 32611, USA;
| | - Tiago Machuca
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
| | - Zhao-Jun Liu
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Omaida C. Velazquez
- DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (F.A.V.); (C.T.H.); (H.S.); (A.R.); (Y.O.); (T.M.)
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Sarker DB, Xue Y, Mahmud F, Jocelyn JA, Sang QXA. Interconversion of Cancer Cells and Induced Pluripotent Stem Cells. Cells 2024; 13:125. [PMID: 38247819 PMCID: PMC10814385 DOI: 10.3390/cells13020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
Cancer cells, especially cancer stem cells (CSCs), share many molecular features with induced pluripotent stem cells (iPSCs) that enable the derivation of induced pluripotent cancer cells by reprogramming malignant cells. Conversely, normal iPSCs can be converted into cancer stem-like cells with the help of tumor microenvironment components and genetic manipulation. These CSC models can be utilized in oncogenic initiation and progression studies, understanding drug resistance, and developing novel therapeutic strategies. This review summarizes the role of pluripotency factors in the stemness, tumorigenicity, and therapeutic resistance of cancer cells. Different methods to obtain iPSC-derived CSC models are described with an emphasis on exposure-based approaches. Culture in cancer cell-conditioned media or cocultures with cancer cells can convert normal iPSCs into cancer stem-like cells, aiding the examination of processes of oncogenesis. We further explored the potential of reprogramming cancer cells into cancer-iPSCs for mechanistic studies and cancer dependencies. The contributions of genetic, epigenetic, and tumor microenvironment factors can be evaluated using these models. Overall, integrating iPSC technology into cancer stem cell research holds significant promise for advancing our knowledge of cancer biology and accelerating the development of innovative and tailored therapeutic interventions.
Collapse
Affiliation(s)
- Drishty B. Sarker
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA; (D.B.S.); (Y.X.); (F.M.); (J.A.J.)
| | - Yu Xue
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA; (D.B.S.); (Y.X.); (F.M.); (J.A.J.)
| | - Faiza Mahmud
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA; (D.B.S.); (Y.X.); (F.M.); (J.A.J.)
| | - Jonathan A. Jocelyn
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA; (D.B.S.); (Y.X.); (F.M.); (J.A.J.)
| | - Qing-Xiang Amy Sang
- Department of Chemistry and Biochemistry, Florida State University, Tallahassee, FL 32306-4390, USA; (D.B.S.); (Y.X.); (F.M.); (J.A.J.)
- Institute of Molecular Biophysics, Florida State University, Tallahassee, FL 32306-4380, USA
| |
Collapse
|
6
|
Xia L, Zhang T, Yao J, Lu K, Hu Z, Gu X, Chen Y, Qin S, Leng W. Fibromodulin overexpression drives oral squamous cell carcinoma via activating downstream EGFR signaling. iScience 2023; 26:108201. [PMID: 37965134 PMCID: PMC10641260 DOI: 10.1016/j.isci.2023.108201] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Accumulating evidence has shown that fibromodulin (FMOD) plays a pivotal role in tumorigenesis and metastasis. However, the biological function of FMOD in oral squamous cell carcinoma (OSCC) remains largely unclear to date. In this study, we confirmed that FMOD was overexpressed and showed a significant association with malignant progression and lymph node metastasis in OSCC. Depletion of FMOD inhibited OSCC proliferation and metastasis in vitro and in vivo. RNA sequencing, western blotting, and rescue assays verified that FMOD exerted oncogenic roles in OSCC via activation of EGFR signaling. In addition, FMOD was proved to be a putative target gene of miR-338-3p. Taken together, FMOD overexpression due to the reduced level of miR-338-3p promotes OSCC by activating EGFR signaling. Our findings provide direct evidence that targeting FMOD could be a promising therapeutic strategy for OSCC patients.
Collapse
Affiliation(s)
- Lingyun Xia
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Tianshu Zhang
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Juncheng Yao
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Kaitian Lu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Ziqiu Hu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| | - Xinsheng Gu
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Yongji Chen
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Shanshan Qin
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Weidong Leng
- Department of Stomatology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, P.R. China
- Institute of Oral Diseases, School of Dentistry, Hubei University of Medicine, Shiyan 442000, P.R. China
| |
Collapse
|
7
|
Liang Y, Zhou Y, Wang J, He Y. Downregulation of fibromodulin attenuates inflammatory signaling and atrial fibrosis in spontaneously hypertensive rats with atrial fibrillation via inhibiting TLR4/NLRP3 signaling pathway. Immun Inflamm Dis 2023; 11:e1003. [PMID: 37904680 PMCID: PMC10604567 DOI: 10.1002/iid3.1003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/05/2023] [Accepted: 08/19/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Myocardial fibrosis is an important factor in the induction and maintenance of atrial fibrillation (AF). Fibromodulin (FMOD) promotes fibrotic gene expression. However, its specific role in spontaneously hypertensive rats (SHR)-AF remains unclear. METHODS We analyzed FMOD mRNA and protein expression in rat atrial tissues using RT-qPCR, Western blot analysis, and immunohistochemistry. Histopathological examination of atrial tissues was performed using hematoxylin and eosin (H&E), Masson's trichrome, and Picrosirius red staining. The levels of inflammatory and fibrosis-related proteins were measured using Western blot analysis. RESULTS FMOD relative mRNA and protein expression levels were notably upregulated in atrial tissues of both AF groups (normal-AF and SHR-AF groups) than that in atrial tissues of the no-AF group (normal and SHR group). This effect was particularly pronounced in the SHR-AF group. Pathological changes revealed that the extracellular matrix, collagen, collagen fibers, and left atrial diameter were notably increased in the atrial tissues from the SHR-AF group compared to those in the atrial tissues from the SHR group, whereas the left ventricular fractional shortening and left ventricular ejection fraction were notably lower. Expression of TLR4, MyD88, NLRP3, TGF-β1, collagen I, and collagen II mRNA were clearly higher in atrial tissues from the SHR-AF group than in those from the SHR group. Protein levels of TLR4, MyD88, NLRP3, Cleavage-Caspase-1, Cleavage-IL-1β, TGF-β1, p-Smad2, collagen I, and collagen II were clearly higher in atrial tissues from the SHR-AF group than in those from the SHR group. FMOD knockdown inhibited atrial fibrosis, collagen accumulation, and the TLR4/MyD88/NLRP3 signaling pathway. CONCLUSION Downregulation of FMOD attenuated inflammatory signaling and atrial fibrosis in SHR-AF by inhibiting the TLR4/NLRP3 signaling pathway. Therefore, FMOD may be a promising therapeutic target in AF.
Collapse
Affiliation(s)
- Yuming Liang
- Department of CardiologyJiangbin Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Yun Zhou
- Department of CardiologyJiangbin Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Jialin Wang
- Health Management CenterThe People's Hospital of Guangxi Zhuang Autonomous RegionNanningChina
| | - Yan He
- Department of Geriatrics CardiologyFirst Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
8
|
Wang P, Meng X, Xue J, Fan C, Wang J. Genome-wide analysis for nanofiber induced global gene expression profile: A study in MC3T3-E1 cells by RNA-Seq. Colloids Surf B Biointerfaces 2023; 223:113143. [PMID: 36682297 DOI: 10.1016/j.colsurfb.2023.113143] [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: 11/21/2022] [Revised: 01/01/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Nanofibers are one of the attractive biomaterials that can provide unique environments to direct cell behaviors. However, how nanofiber structure affects the global gene expression of laden cells remains unclear. Herein, high-throughput mRNA sequencing (RNA-seq) is applied to analyze the transcriptome of the MC3T3-E1 cells (a model osteoblast cell line) cultured on electrospun nanofibers. The cell-adhesive poly(L-lactide) nanofibers and membranes are developed by the mussel-inspired coating of gelatin-dopamine conjugate under H2O2-mediated oxidation. The MC3T3-E1 cells cultured on nanofibers exhibit elongated morphology and increased proliferation compared with those on membranes. The differences in global gene expression profiles are determined by RNA-seq, in which 905 differentially expressed genes (DEGs) are identified. Significantly, the DEGs related to cytoskeleton, promotion of cell cycle progression, cell adhesion, and cell proliferation, are higher expressed in the cells on nanofibers, while the DEGs involved in cell-cycle arrest and osteoblast mineralization are up-regulated in the cells on membranes. This study elucidates the roles of nanofiber structure in affecting gene expression of laden cells at the whole transcriptome level, and it will lay the foundation for understanding nanofiber-guided cell behaviors.
Collapse
Affiliation(s)
- Peiyan Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China
| | - Xinyue Meng
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China
| | - Junqiang Xue
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266000, Shandong, PR China
| | - Changjiang Fan
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China.
| | - Jianxun Wang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong, PR China; Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao 266021, Shandong, PR China.
| |
Collapse
|
9
|
Denervation during mandibular distraction osteogenesis results in impaired bone formation. Sci Rep 2023; 13:2097. [PMID: 36747028 PMCID: PMC9902545 DOI: 10.1038/s41598-023-27921-9] [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: 09/08/2022] [Accepted: 01/10/2023] [Indexed: 02/08/2023] Open
Abstract
Mandibular distraction osteogenesis (DO) is mediated by skeletal stem cells (SSCs) in mice, which enact bone regeneration via neural crest re-activation. As peripheral nerves are essential to progenitor function during development and in response to injury, we questioned if denervation impairs mandibular DO. C57Bl6 mice were divided into two groups: DO with a segmental defect in the inferior alveolar nerve (IAN) at the time of mandibular osteotomy ("DO Den") and DO with IAN intact ("DO Inn"). DO Den demonstrated significantly reduced histological and radiological osteogenesis relative to DO Inn. Denervation preceding DO results in reduced SSC amplification and osteogenic potential in mice. Single cell RNA sequencing analysis revealed that there was a predominance of innervated SSCs in clusters dominated by pathways related to bone formation. A rare human patient specimen was also analyzed and suggested that histological, radiological, and transcriptional alterations seen in mouse DO may be conserved in the setting of denervated human mandible distraction. Fibromodulin (FMOD) transcriptional and protein expression were reduced in denervated relative to innervated mouse and human mandible regenerate. Finally, when exogenous FMOD was added to DO-Den and DO-Inn SSCs undergoing in vitro osteogenic differentiation, the osteogenic potential of DO-Den SSCs was increased in comparison to control untreated DO-Den SSCs, modeling the superior osteogenic potential of DO-Inn SSCs.
Collapse
|
10
|
Abstract
Fibromodulin (FMOD) is an archetypal member of the class II small leucine-rich proteoglycan family. By directly binding to extracellular matrix structural components, such as collagen and lysyl oxidase, FMOD regulates collagen cross-linking, packing, assembly, and fibril architecture via a multivalent interaction. Meanwhile, as a pluripotent molecule, FMOD acts as a ligand of various cytokines and growth factors, especially those belonging to the transforming growth factor (TGF) β superfamily, by interacting with the corresponding signaling molecules involved in cell adhesion, spreading, proliferation, migration, invasion, differentiation, and metastasis. Consequently, FMOD exhibits promigratory, proangiogenic, anti-inflammatory, and antifibrogenic properties and plays essential roles in cell fate determination and maturation, progenitor cell recruitment, and tissue regeneration. The multifunctional nature of FMOD thus enables it to be a promising therapeutic agent for a broad repertoire of diseases, including but not limited to arthritis, temporomandibular joint disorders, caries, and fibrotic diseases among different organs, as well as to be a regenerative medicine candidate for skin, muscle, and tendon injuries. Moreover, FMOD is also considered a marker for tumor diagnosis and prognosis prediction and a potential target for cancer treatment. Furthermore, FMOD itself is sufficient to reprogram somatic cells into a multipotent state, creating a safe and efficient cell source for various tissue reconstructions and thus opening a new avenue for regenerative medicine. This review focuses on the recent preclinical efforts bringing FMOD research and therapies to the forefront. In addition, a contemporary understanding of the mechanism underlying FMOD's function, particularly its interaction with TGFβ superfamily members, is also discussed at the molecular level to aid the discovery of novel FMOD-based treatments.
Collapse
Affiliation(s)
- Z. Zheng
- David Geffen School of Medicine,
University of California, Los Angeles, CA, USA
- School of Dentistry, University of
California, Los Angeles, CA, USA
| | - H.S. Granado
- Department of Orthodontics, School of
Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - C. Li
- Department of Orthodontics, School of
Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Xu X, Zhang Y, Ha P, Chen Y, Li C, Yen E, Bai Y, Chen R, Wu BM, Da Lio A, Ting K, Soo C, Zheng Z. A novel injectable fibromodulin-releasing granular hydrogel for tendon healing and functional recovery. Bioeng Transl Med 2023; 8:e10355. [PMID: 36684085 PMCID: PMC9842059 DOI: 10.1002/btm2.10355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/06/2022] [Accepted: 05/07/2022] [Indexed: 01/25/2023] Open
Abstract
A crucial component of the musculoskeletal system, the tendon is one of the most commonly injured tissues in the body. In severe cases, the ruptured tendon leads to permanent dysfunction. Although many efforts have been devoted to seeking a safe and efficient treatment for enhancing tendon healing, currently existing treatments have not yet achieved a major clinical improvement. Here, an injectable granular hyaluronic acid (gHA)-hydrogel is engineered to deliver fibromodulin (FMOD)-a bioactive extracellular matrix (ECM) that enhances tenocyte mobility and optimizes the surrounding ECM assembly for tendon healing. The FMOD-releasing granular HA (FMOD/gHA)-hydrogel exhibits unique characteristics that are desired for both patients and health providers, such as permitting a microinvasive application and displaying a burst-to-sustained two-phase release of FMOD, which leads to a prompt FMOD delivery followed by a constant dose-maintaining period. Importantly, the generated FMOD-releasing granular HA hydrogel significantly augmented tendon-healing in a fully-ruptured rat's Achilles tendon model histologically, mechanically, and functionally. Particularly, the breaking strength of the wounded tendon and the gait performance of treated rats returns to the same normal level as the healthy controls. In summary, a novel effective FMOD/gHA-hydrogel is developed in response to the urgent demand for promoting tendon healing.
Collapse
Affiliation(s)
- Xue Xu
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yulong Zhang
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Pin Ha
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| | - Yao Chen
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chenshuang Li
- Department of OrthodonticsSchool of Dental Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Emily Yen
- Arcadia High SchoolArcadiaCaliforniaUSA
| | - Yuxing Bai
- Department of OrthodonticsBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Renji Chen
- Department of Oral and Maxillofacial Plastic and Traumatic SurgeryBeijing Stomatological Hospital of Capital Medical UniversityBeijingChina
| | - Benjamin M. Wu
- School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Andrew Da Lio
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
| | - Kang Ting
- Forsyth Research InstituteHarvard UniversityCambridgeMassachusettsUSA
- Samueli School of EngineeringUniversity of CaliforniaLos AngelesCaliforniaUSA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic SurgeryThe Orthopaedic Hospital Research Center, University of CaliforniaLos AngelesCaliforniaUSA
| | - Zhong Zheng
- Division of Plastic and Reconstructive SurgeryDavid Geffen School of Medicine, University of CaliforniaLos AngelesCaliforniaUSA
- Division of Growth and DevelopmentSchool of Dentistry, University of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
12
|
Halasi M, Grinstein M, Adini A, Adini I. Fibromodulin Ablation Exacerbates the Severity of Acute Colitis. J Inflamm Res 2022; 15:4515-4526. [PMID: 35966006 PMCID: PMC9374093 DOI: 10.2147/jir.s366290] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/17/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Epidemiological studies have associated pigment production with protection against certain human diseases. In contrast to African Americans, European descendants are more likely to suffer from angiogenesis-dependent and inflammatory diseases, such as wet age-related macular degeneration (ARMD) and ulcerative colitis (UC), respectively. Methods In a mouse model of dextran sulfate sodium (DSS)-induced acute colitis, the effect of fibromodulin (FMOD) depletion was examined on colitis severity. Results In this study, albino mice that produce high levels of FMOD developed less severe acute colitis compared with mice lacking in FMOD as assessed by clinical symptoms and histopathological changes. FMOD depletion affected the expression of tight junction proteins, contributing to the destruction of the epithelial barrier. Furthermore, this study revealed a stronger inflammatory response after DSS treatment in the absence of FMOD, where FMOD depletion led to an increase in activated T cells, plasmacytoid dendritic cells (pDCs), and type I interferon (IFN) production. Discussion These findings point to FMOD as a potential biomarker of disease severity in UC among light-skinned individuals of European descent.
Collapse
Affiliation(s)
- Marianna Halasi
- Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Mor Grinstein
- Department of Medicine, Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Avner Adini
- Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Irit Adini
- Department of Surgery, Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Oria VO, Zhang H, Zito CR, Rane CK, Ma XY, Provance OK, Tran TT, Adeniran A, Kluger Y, Sznol M, Bosenberg MW, Kluger HM, Jilaveanu LB. Coupled fibromodulin and SOX2 signaling as a critical regulator of metastatic outgrowth in melanoma. Cell Mol Life Sci 2022; 79:377. [PMID: 35737114 PMCID: PMC9226089 DOI: 10.1007/s00018-022-04364-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 04/19/2022] [Accepted: 05/08/2022] [Indexed: 11/26/2022]
Abstract
We aimed to study mechanisms controlling metastatic outgrowth of melanoma into clinically relevant lesions, a critical process responsible for the majority of melanoma deaths. To this end, we developed novel in vivo models and identified molecular events that can be ascribed to their distinct phenotypes, indolent or highly metastatic. Induction of a proliferative state at distant sites was associated with high levels of the stem-like/progenitor marker, SOX2, and required the upregulation of FMOD, an extracellular matrix component, which modulates tumor-stroma interactions. Functional studies revealed a possible link between FMOD and SOX2; dual FMOD and SOX2 silencing nearly abolished brain metastasis and had a similar effect on distant metastasis to other sites. Our in vitro data suggests that FMOD and SOX2 cooperation plays an important role in tumor vasculogenic mimicry. Furthermore, we found that FMOD and SOX2 functional roles might converge at the activation of transcriptional co-factors YAP and TAZ, possibly via crosstalk with the tumor suppressor Hippo pathway. Finally, high expression of both genes in patient specimens predicted early development of brain metastasis. Thus, our study identifies FMOD and SOX2 cooperation as a novel regulatory mechanism that might be linked functionally to melanoma metastatic competence.
Collapse
Affiliation(s)
- Victor O Oria
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Hongyi Zhang
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Christopher R Zito
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
- Department of Biology, School of Arts, Sciences, Business, and Education, University of Saint Joseph, West Hartford, CT, USA
| | - Chetan K Rane
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Xian-Yong Ma
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Olivia K Provance
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Thuy T Tran
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Adebowale Adeniran
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Mario Sznol
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Marcus W Bosenberg
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Harriet M Kluger
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA
| | - Lucia B Jilaveanu
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, 333 Cedar Street, SHM234E, New Haven, CT, 06520, USA.
| |
Collapse
|
14
|
von Mentzer U, Corciulo C, Stubelius A. Biomaterial Integration in the Joint: Pathological Considerations, Immunomodulation, and the Extracellular Matrix. Macromol Biosci 2022; 22:e2200037. [PMID: 35420256 DOI: 10.1002/mabi.202200037] [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: 02/27/2022] [Revised: 03/30/2022] [Indexed: 11/08/2022]
Abstract
Defects of articular joints are becoming an increasing societal burden due to a persistent increase in obesity and aging. For some patients suffering from cartilage erosion, joint replacement is the final option to regain proper motion and limit pain. Extensive research has been undertaken to identify novel strategies enabling earlier intervention to promote regeneration and cartilage healing. With the introduction of decellularized extracellular matrix (dECM), researchers have tapped into the potential for increased tissue regeneration by designing biomaterials with inherent biochemical and immunomodulatory signals. Compared to conventional and synthetic materials, dECM-based materials invoke a reduced foreign body response. It is therefore highly beneficial to understand the interplay of how these native tissue-based materials initiate a favorable remodeling process by the immune system. Yet, such an understanding also demands increasing considerations of the pathological environment and remodeling processes, especially for materials designed for early disease intervention. This knowledge would avoid rejection and help predict complications in conditions with inflammatory components such as arthritides. This review outlines general issues facing biomaterial integration and emphasizes the importance of tissue-derived macromolecular components in regulating essential homeostatic, immunological, and pathological processes to increase biomaterial integration for patients suffering from joint degenerative diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Ula von Mentzer
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| | - Carmen Corciulo
- Centre for Bone and Arthritis Research, Department of Rheumatology and Inflammation, Sahlgrenska Academy at the University of Gothenburg, Guldhedsgatan 10A, Gothenburg, 41296, Sweden
| | - Alexandra Stubelius
- Division of Chemical Biology, Department of Biology and Biological Engineering, Chalmers University of Technology, Kemivägen 10, Gothenburg, 41296, Sweden
| |
Collapse
|
15
|
Li C, Mills Z, Zheng Z. Novel cell sources for bone regeneration. MedComm (Beijing) 2021; 2:145-174. [PMID: 34766140 PMCID: PMC8491221 DOI: 10.1002/mco2.51] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
A plethora of both acute and chronic conditions, including traumatic, degenerative, malignant, or congenital disorders, commonly induce bone disorders often associated with severe persisting pain and limited mobility. Over 1 million surgical procedures involving bone excision, bone grafting, and fracture repair are performed each year in the U.S. alone, resulting in immense levels of public health challenges and corresponding financial burdens. Unfortunately, the innate self-healing capacity of bone is often inadequate for larger defects over a critical size. Moreover, as direct transplantation of committed osteoblasts is hindered by deficient cell availability, limited cell spreading, and poor survivability, an urgent need for novel cell sources for bone regeneration is concurrent. Thanks to the development in stem cell biology and cell reprogramming technology, many multipotent and pluripotent cells that manifest promising osteogenic potential are considered the regenerative remedy for bone defects. Considering these cells' investigation is still in its relative infancy, each of them offers their own particular challenges that must be conquered before the large-scale clinical application.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Zane Mills
- College of DentistryUniversity of OklahomaOklahoma CityOklahomaUSA
| | - Zhong Zheng
- Division of Growth and Development, School of DentistryUniversity of CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, David Geffen School of MedicineUniversity of CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
16
|
Kipper FC, Angolano C, Vissapragada R, Contreras MA, Moore J, Bhasin M, Ferran C, Thomas AJ. Embryonic periventricular endothelial cells demonstrate a unique pro-neurodevelopment and anti-inflammatory gene signature. Sci Rep 2020; 10:20393. [PMID: 33230288 PMCID: PMC7683543 DOI: 10.1038/s41598-020-77297-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/02/2020] [Indexed: 01/08/2023] Open
Abstract
Brain embryonic periventricular endothelial cells (PVEC) crosstalk with neural progenitor cells (NPC) promoting mutual proliferation, formation of tubular-like structures in the former and maintenance of stemness in the latter. To better characterize this interaction, we conducted a comparative transcriptome analysis of mouse PVEC vs. adult brain endothelial cells (ABEC) in mono-culture or NPC co-culture. We identified > 6000 differentially expressed genes (DEG), regardless of culture condition. PVEC exhibited a 30-fold greater response to NPC than ABEC (411 vs. 13 DEG). Gene Ontology (GO) analysis of DEG that were higher or lower in PVEC vs. ABEC identified "Nervous system development" and "Response to Stress" as the top significantly different biological process, respectively. Enrichment in canonical pathways included HIF1A, FGF/stemness, WNT signaling, interferon signaling and complement. Solute carriers (SLC) and ABC transporters represented an important subset of DEG, underscoring PVEC's implication in blood-brain barrier formation and maintenance of nutrient-rich/non-toxic environment. Our work characterizes the gene signature of PVEC and their important partnership with NPC, underpinning their unique role in maintaining a healthy neurovascular niche, and in supporting brain development. This information may pave the way for additional studies to explore their therapeutic potential in neuro-degenerative diseases, such as Alzheimer's and Parkinson's disease.
Collapse
Affiliation(s)
- Franciele Cristina Kipper
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Cleide Angolano
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Ravi Vissapragada
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Discipline of Surgery, College of Medicine and Public Health, Flinders University, Adelaide, 5042, SA, Australia
| | - Mauricio A Contreras
- Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Justin Moore
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Manoj Bhasin
- BIDMC Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Division of Vascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA
- Harvard Medical School, Boston, MA, 02215, USA
| | - Ajith J Thomas
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, MA, 02215, USA.
- Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
17
|
Yang P, Li C, Lee M, Marzvanyan A, Zhao Z, Ting K, Soo C, Zheng Z. Photopolymerizable Hydrogel-Encapsulated Fibromodulin-Reprogrammed Cells for Muscle Regeneration. Tissue Eng Part A 2020; 26:1112-1122. [PMID: 32323608 PMCID: PMC7580647 DOI: 10.1089/ten.tea.2020.0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/14/2020] [Indexed: 02/05/2023] Open
Abstract
A central challenge in tissue engineering is obtaining a suitable cell type with a capable delivery vehicle to replace or repair damaged or diseased tissues with tissue mimics. Notably, for skeletal muscle tissue engineering, given the inadequate availability and regenerative capability of endogenous myogenic progenitor cells as well as the tumorigenic risks presented by the currently available pluri- and multipotent stem cells, seeking a safe regenerative cell source is urgently demanded. To conquer this problem, we previously established a novel reprogramming technology that can generate multipotent cells from dermal fibroblasts using a single protein, fibromodulin (FMOD). The yield FMOD-reprogrammed (FReP) cells exhibit exceeding myogenic capability without tumorigenic risk, making them a promising and safe cell source for skeletal muscle establishment. In addition to using the optimal cell for implantation, it is equally essential to maintain cellular localization and retention in the recipient tissue environment for critical-sized muscle tissue establishment. In this study, we demonstrate that the photopolymerizable methacrylated glycol chitosan (MeGC)/type I collagen (ColI)-hydrogel provides a desirable microenvironment for encapsulated FReP cell survival, spreading, extension, and formation of myotubes in the hydrogel three-dimensionally in vitro, without undesired osteogenic, chondrogenic, or tenogenic differentiation. Furthermore, gene profiling revealed a paired box 7 (PAX7) → myogenic factor 5 (MYF5) → myogenic determination 1 (MYOD1) → myogenin (MYOG) → myosin cassette elevation in the encapsulated FReP cells during myogenic differentiation, which is similar to that of the predominant driver of endogenous skeletal muscle regeneration, satellite cells. These findings constitute the evidence that the FReP cell-MeGC/ColI-hydrogel construct is a promising tissue engineering mimic for skeletal muscle generation in vitro, and thus possesses the extraordinary potential for further in vivo validation.
Collapse
Affiliation(s)
- Pu Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Chenshuang Li
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California, USA
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Min Lee
- Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, California, USA
| | - Anna Marzvanyan
- A. T. Still University School of Osteopathic Medicine in Arizona, Mesa, Arizona, USA
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, People's Republic of China
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California, USA
| | - Chia Soo
- UCLA Division of Plastic Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California, USA
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, Dental and Craniofacial Research Institute, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
18
|
Yin H, Cui C, Han S, Chen Y, Zhao J, He H, Li D, Zhu Q. Fibromodulin Modulates Chicken Skeletal Muscle Development via the Transforming Growth Factor-β Signaling Pathway. Animals (Basel) 2020; 10:ani10091477. [PMID: 32842630 PMCID: PMC7552301 DOI: 10.3390/ani10091477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/18/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Fibromodulin (Fmod) plays critical roles in skeletal muscle development and maintenance, but the roles of Fmod in skeletal muscle atrophy and development in chickens are unclear. Here, we demonstrate that Fmod plays important roles in the differentiation and atrophy of chicken skeletal muscle by regulating the transforming growth factor-β signaling pathway. These results suggest that Fmod plays important roles in skeletal muscle growth and development in chickens. Abstract Fibromodulin (Fmod), which is an extracellular matrix protein, belongs to the extracellular matrix small-leucine-rich proteoglycan family. Fmod is abundantly expressed in muscles and connective tissues and is involved in biological regulation processes, including cell apoptosis, cell adhesion, and modulation of cytokine activity. Fmod is the main regulator of myostatin, which controls the development of muscle cells, but its regulatory path is unknown. Chicken models are ideal for studying embryonic skeletal muscle development; therefore, to investigate the mechanism of Fmod in muscle development, Fmod-silenced and Fmod-overexpressed chicken myoblasts were constructed. The results showed that Fmod plays a positive role in differentiation by detecting the expression of myogenic differentiation markers, immunofluorescence of MyHC protein, and myotube formation in myoblasts. Fmod regulates expression of atrophy-related genes to alleviate muscle atrophy, which was confirmed by histological analysis of breast muscles in Fmod-modulated chicks in vivo. Additionally, genes differentially expressed between Fmod knockdown and normal myoblasts were enriched in the signaling pathway of transforming growth factor β (TGF-β). Both Fmod-silenced and Fmod-overexpressed myoblasts regulated the expression of TGFBR1 and p-Smad3. Thus, Fmod can promote differentiation but not proliferation of myoblasts by regulating the TGF-β signaling pathway, which may serve a function in muscular atrophy.
Collapse
|
19
|
Walimbe T, Panitch A. Proteoglycans in Biomedicine: Resurgence of an Underexploited Class of ECM Molecules. Front Pharmacol 2020; 10:1661. [PMID: 32082161 PMCID: PMC7000921 DOI: 10.3389/fphar.2019.01661] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 12/18/2019] [Indexed: 01/02/2023] Open
Abstract
Proteoglycans have emerged as biomacromolecules with important roles in matrix remodeling, homeostasis, and signaling in the past two decades. Due to their negatively charged glycosaminoglycan chains as well as distinct core protein structures, they interact with a variety of molecules, including matrix proteins, growth factors, cytokines and chemokines, pathogens, and enzymes. This led to the dawn of glycan therapies in the 20th century, but this research was quickly overshadowed by readily available DNA and protein-based therapies. The recent development of recombinant technology and advances in our understanding of proteoglycan function have led to a resurgence of these molecules as potential therapeutics. This review focuses on the recent preclinical efforts that are bringing proteoglycan research and therapies back to the forefront. Examples of studies using proteoglycan cores and mimetics have also been included to give the readers a perspective on the wide-ranging and extensive applications of these versatile molecules. Collectively, these advances are opening new avenues for targeting diseases at a molecular level, and providing avenues for the development of new and exciting treatments in regenerative medicine.
Collapse
Affiliation(s)
- Tanaya Walimbe
- Laboratory of Engineered Therapeutics, Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Alyssa Panitch
- Laboratory of Engineered Therapeutics, Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| |
Collapse
|
20
|
Pang X, Dong N, Zheng Z. Small Leucine-Rich Proteoglycans in Skin Wound Healing. Front Pharmacol 2020; 10:1649. [PMID: 32063855 PMCID: PMC6997777 DOI: 10.3389/fphar.2019.01649] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022] Open
Abstract
Healing of cutaneous wounds is a complex and well-coordinated process requiring cooperation among multiple cells from different lineages and delicately orchestrated signaling transduction of a diversity of growth factors, cytokines, and extracellular matrix (ECM) at the wound site. Most skin wound healing in adults is imperfect, characterized by scar formation which results in significant functional and psychological sequelae. Thus, the reconstruction of the damaged skin to its original state is of concern to doctors and scientists. Beyond the traditional treatments such as corticosteroid injection and radiation therapy, several growth factors or cytokines-based anti-scarring products are being or have been tested in clinical trials to optimize skin wound healing. Unfortunately, all have been unsatisfactory to date. Currently, accumulating evidence suggests that the ECM not only functions as the structural component of the tissue but also actively modulates signal transduction and regulates cellular behaviors, and thus, ECM should be considered as an alternative target for wound management pharmacotherapy. Of particular interest are small leucine-rich proteoglycans (SLRPs), a group of the ECM, which exist in a wide range of connecting tissues, including the skin. This manuscript summarizes the most current knowledge of SLRPs regarding their spatial-temporal expression in the skin, as well as lessons learned from the genetically modified animal models simulating human skin pathologies. In this review, particular focus is given on the diverse roles of SLRP in skin wound healing, such as anti-inflammation, pro-angiogenesis, pro-migration, pro-contraction, and orchestrate transforming growth factor (TGF)β signal transduction, since cumulative investigations have indicated their therapeutic potential on reducing scar formation in cutaneous wounds. By conducting this review, we intend to gain insight into the potential application of SLRPs in cutaneous wound healing management which may pave the way for the development of a new generation of pharmaceuticals to benefit the patients suffering from skin wounds and their sequelae.
Collapse
Affiliation(s)
- Xiaoxiao Pang
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, China
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Nuo Dong
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
21
|
Targeting cell plasticity for regeneration: From in vitro to in vivo reprogramming. Adv Drug Deliv Rev 2020; 161-162:124-144. [PMID: 32822682 DOI: 10.1016/j.addr.2020.08.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/14/2022]
Abstract
The discovery of induced pluripotent stem cells (iPSCs), reprogrammed to pluripotency from somatic cells, has transformed the landscape of regenerative medicine, disease modelling and drug discovery pipelines. Since the first generation of iPSCs in 2006, there has been enormous effort to develop new methods that increase reprogramming efficiency, and obviate the need for viral vectors. In parallel to this, the promise of in vivo reprogramming to convert cells into a desired cell type to repair damage in the body, constitutes a new paradigm in approaches for tissue regeneration. This review article explores the current state of reprogramming techniques for iPSC generation with a specific focus on alternative methods that use biophysical and biochemical stimuli to reduce or eliminate exogenous factors, thereby overcoming the epigenetic barrier towards vector-free approaches with improved clinical viability. We then focus on application of iPSC for therapeutic approaches, by giving an overview of ongoing clinical trials using iPSCs for a variety of health conditions and discuss future scope for using materials and reagents to reprogram cells in the body.
Collapse
|
22
|
Li C, Ha P, Jiang W, Haveles CS, Zheng Z, Zou M. Fibromodulin - A New Target of Osteoarthritis Management? Front Pharmacol 2019; 10:1475. [PMID: 31920661 PMCID: PMC6927287 DOI: 10.3389/fphar.2019.01475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/13/2019] [Indexed: 12/15/2022] Open
Affiliation(s)
- Chenshuang Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States.,Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Pin Ha
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Wenlu Jiang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Christos S Haveles
- David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, United States
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, United States
| | - Min Zou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Department of Orthodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
23
|
Zheng Z, Li C, Ha P, Chang GX, Yang P, Zhang X, Kim JK, Jiang W, Pang X, Berthiaume EA, Mills Z, Haveles CS, Chen E, Ting K, Soo C. CDKN2B upregulation prevents teratoma formation in multipotent fibromodulin-reprogrammed cells. J Clin Invest 2019; 129:3236-3251. [PMID: 31305260 PMCID: PMC6668700 DOI: 10.1172/jci125015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 05/21/2019] [Indexed: 12/11/2022] Open
Abstract
Tumorigenicity is a well-documented risk to overcome for pluripotent or multipotent cell applications in regenerative medicine. To address the emerging demand for safe cell sources in tissue regeneration, we established a novel, protein-based reprogramming method that does not require genome integration or oncogene activation to yield multipotent fibromodulin (FMOD)-reprogrammed (FReP) cells from dermal fibroblasts. When compared with induced pluripotent stem cells (iPSCs), FReP cells exhibited a superior capability for bone and skeletal muscle regeneration with markedly less tumorigenic risk. Moreover, we showed that the decreased tumorigenicity of FReP cells was directly related to an upregulation of cyclin-dependent kinase inhibitor 2B (CDKN2B) expression during the FMOD reprogramming process. Indeed, sustained suppression of CDKN2B resulted in tumorigenic, pluripotent FReP cells that formed teratomas in vivo that were indistinguishable from iPSC-derived teratomas. These results highlight the pivotal role of CDKN2B in cell fate determination and tumorigenic regulation and reveal an alternative pluripotent/multipotent cell reprogramming strategy that solely uses FMOD protein.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, and
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, and
| | - Pin Ha
- Division of Growth and Development, School of Dentistry, and
| | - Grace X. Chang
- David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Pu Yang
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, and
| | - Jong Kil Kim
- Division of Growth and Development, School of Dentistry, and
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, and
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoxiao Pang
- Division of Growth and Development, School of Dentistry, and
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatology Hospital of Chongqing Medical University, Chongqing, China
| | | | - Zane Mills
- Department of Ecology and Evolutionary Biology, and
| | | | - Eric Chen
- Division of Growth and Development, School of Dentistry, and
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, and
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, UCLA, Los Angeles, California, USA
| |
Collapse
|
24
|
Pourhanifeh MH, Mohammadi R, Noruzi S, Hosseini SA, Fanoudi S, Mohamadi Y, Hashemzehi M, Asemi Z, Mirzaei HR, Salarinia R, Mirzaei H. The role of fibromodulin in cancer pathogenesis: implications for diagnosis and therapy. Cancer Cell Int 2019; 19:157. [PMID: 31198406 PMCID: PMC6558739 DOI: 10.1186/s12935-019-0870-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/27/2019] [Indexed: 01/09/2023] Open
Abstract
Fibromodulin (FMOD) is known as one of very important extracellular matrix small leucine-rich proteoglycans. This small leucine-rich proteoglycan has critical roles in the extracellular matrix organization and necessary for repairing of tissue in many organs. Given that the major task of FMOD is the modulation of collagen fibrillogenesis. However, recently observed that FMOD plays very important roles in the modulation of a variety of pivotal biological processes including angiogenesis, regulation of TGF-β activity, and differentiation of human fibroblasts into pluripotent cells, inflammatory mechanisms, apoptosis and metastatic related phenotypes. Besides these roles, FMOD has been considered as a new tumor-related antigen in some malignancies such as lymphoma, leukemia, and leiomyoma. Taken together, these findings proposed that FMOD could be introduced as diagnostic and therapeutic biomarkers in treatment of various cancers. Herein, for first time, we highlighted the various roles of FMOD in the cancerous conditions. Moreover, we summarized the diagnostic and therapeutic applications of FMOD in cancer therapy.
Collapse
Affiliation(s)
- Mohammad Hossein Pourhanifeh
- 1Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Rezvan Mohammadi
- 2Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Somaye Noruzi
- 2Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyede Atefe Hosseini
- 2Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sahar Fanoudi
- 3Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yousef Mohamadi
- 4Department of Anatomy, Faculty of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Milad Hashemzehi
- Iranshahr University of Medical Sciences, Iranshahr, Iran.,6Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zatollah Asemi
- 1Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Reza Mirzaei
- 7Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Salarinia
- 2Department of Medical Biotechnology and Molecular Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hamed Mirzaei
- 1Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
25
|
Knockdown of Fibromodulin Inhibits Proliferation and Migration of RPE Cell via the VEGFR2-AKT Pathway. J Ophthalmol 2018; 2018:5708537. [PMID: 30298106 PMCID: PMC6157207 DOI: 10.1155/2018/5708537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/12/2018] [Indexed: 12/28/2022] Open
Abstract
Purpose Recent research has provided novel insight into the function of fibromodulin (FMOD) in wound healing and angiogenesis. The role of FMOD in initiation of proliferative vitreoretinopathy (PVR) has not been studied. This study investigated the effect of FMOD on human retinal pigment epithelial (RPE) cell, which plays an essential role in the progression of PVR, and the possible mechanisms. Methods Small interfering (si) RNA-based gene transfer technology was used to decrease FMOD expression and to study its effects on RPEs in vitro. Cell Counting Kit-8 assays, transwells, and flow cytometry analysis were used to measure cell proliferation, migration, cell cycle, and apoptosis. Western blot was used to measure expression of vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), extracellular signal-related kinase 1/2 (ERK1/2), and phosphoinositide 3 kinase (PI3K/AKT). Results After transfection of RPEs with a FMOD-specific siRNA, cell proliferation and migration were inhibited to the percentage of 65% ± 5% and 39% ± 10%, respectively, compared to the control group. Depletion of FMOD induced cell cycle arrest and apoptosis in RPE cells. Downregulation of VEGF, VEGFR2, and phosphorylated AKT (p-AKT) were detected in transfected RPEs. Conclusion Depletion of FMOD selectively downregulated the expression of VEGF and VEGFR2 and inhibited the signaling pathway of AKT phosphorylation, which consequently inhibited the proliferation and migration of RPE Cell.
Collapse
|
26
|
Lipman K, Wang C, Ting K, Soo C, Zheng Z. Tendinopathy: injury, repair, and current exploration. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:591-603. [PMID: 29593382 PMCID: PMC5865563 DOI: 10.2147/dddt.s154660] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Both acute and chronic tendinopathy result in high morbidity, requiring management that is often lengthy and expensive. However, limited and conflicting scientific evidence surrounding current management options has presented a challenge when trying to identify the best treatment for tendinopathy. As a result of shortcomings of current treatments, response to available therapies is often poor, resulting in frustration in both patients and physicians. Due to a lack of understanding of basic tendon-cell biology, further scientific investigation is needed in the field for the development of biological solutions. Optimization of new delivery systems and therapies that spatially and temporally mimic normal tendon physiology hold promise for clinical application. This review focuses on the clinical importance of tendinopathy, the structure of healthy tendons, tendon injury, and healing, and a discussion of current approaches for treatment that highlight the need for the development of new nonsurgical interventions.
Collapse
Affiliation(s)
| | - Chenchao Wang
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA.,First Hospital of China Medical University, Shenyang, China.,Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Kang Ting
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, Orthopaedic Hospital Research Center, University of California, Los Angeles, CA, USA
| | - Zhong Zheng
- Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Lee EJ, Jan AT, Baig MH, Ahmad K, Malik A, Rabbani G, Kim T, Lee IK, Lee YH, Park SY, Choi I. Fibromodulin and regulation of the intricate balance between myoblast differentiation to myocytes or adipocyte-like cells. FASEB J 2018; 32:768-781. [PMID: 28974563 DOI: 10.1096/fj.201700665r] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Interactions between myoblasts and the surrounding microenvironment led us to explore the role of fibromodulin (FMOD), an extracellular matrix protein, in the maintenance of myoblast stemness and function. Microarray analysis of FMODkd myoblasts and in silico studies were used to identify the top most differentially expressed genes in FMODkd, and helped establish that FMOD-based regulations of integral membrane protein 2a and clusterin are essential components of the myogenic program. Studies in knockout, obese, and diabetic mouse models helped characterize the operation of a novel FMOD-based regulatory circuit that controls myoblast switching from a myogenic to a lipid accumulation fate. FMOD regulation of myoblasts is an essential part of the myogenic program, and it offers opportunities for the development of therapeutics for the treatment of different muscle diseases.-Lee, E. J., Jan, A. T., Baig, M. H., Ahmad, K., Malik, A., Rabbani, G., Kim, T., Lee, I.-K., Lee, Y. H., Park, S.-Y., Choi, I. Fibromodulin and regulation of the intricate balance between myoblast differentiation to myocytes or adipocyte-like cells.
Collapse
Affiliation(s)
- Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Mohammad Hassan Baig
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Khurshid Ahmad
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Adeel Malik
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, South Korea
| | - Gulam Rabbani
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - Taeyeon Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| | - In-Kyu Lee
- Department of Internal Medicine, School of Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Yong Ho Lee
- Department of Biomedical Science, Catholic University of Daegu, Gyeongsan, South Korea; and
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, South Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
28
|
Hisanaga Y, Suzuki E, Aoki H, Sato M, Saito A, Saito A, Azuma T. Effect of the combined use of enamel matrix derivative and atelocollagen sponge scaffold on osteoblastic differentiation of mouse induced pluripotent stem cells in vitro. J Periodontal Res 2017; 53:240-249. [PMID: 29044527 DOI: 10.1111/jre.12511] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/07/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND AND OBJECTIVE Induced pluripotent stem cells (iPSCs) are a candidate cell source in periodontal regenerative therapy. Enamel matrix derivative (EMD) has been shown to regenerate periodontal tissues, and atelocollagen sponge (ACS) is considered a suitable scaffold or carrier for growth factors. This study aimed to investigate the effect of combined use of EMD and an ACS scaffold on cell behaviors and differentiation of mouse iPSCs (miPSCs) in vitro. MATERIAL AND METHODS Following embryonic body formation from miPSCs, dissociated cells (miPS-EB-derived cells) were seeded onto ACS with or without EMD, and cultured in osteoblast differentiation medium. Scanning electron microscopy and histological analyses were used to assess cell morphology and infiltration within the ACS. Cell viability (metabolism) was determined using an MTS assay, and expression of mRNA of osteoblastic differentiation markers was assessed by quantitative RT -PCR. Alkaline phosphatase (ALP) staining intensity and activity were evaluated. Mineralization was assessed by von Kossa staining, and calcium content was quantitated using the methylxylenol blue method. RESULTS By 24 hours after seeding, miPS-EB-derived cells in both the EMD and control groups had attached to and infiltrated the ACS scaffold. Scanning electron microscopy images revealed that by day 14, many cytoplasmic protrusions and extracellular deposits, suggestive of calcified matrix, were present in the EMD group. There was a time-dependent increase in cell viability up to day 3, but no difference between groups was observed at any time point. The levels expressed of ALP and osterix genes were significantly higher in the EMD group than in the control group. Expression of runt-related transcription factor 2 was increased in the EMD group compared with the control group on day 7. EMD upregulated the expression of bone sialoprotein and osteopontin on day 14, whereas expression of osteocalcin was lower at all time points. The staining intensity and activity of ALP were higher in the EMD group than in the control group. Mineralization levels and calcium contents were significantly higher in the EMD group throughout the observation period. CONCLUSION These data suggest that combining ACS with EMD increases levels of osteoblastic differentiation and mineralization in miPS-EB-derived cells, compared with ACS used alone.
Collapse
Affiliation(s)
- Y Hisanaga
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - E Suzuki
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - H Aoki
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - M Sato
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan
| | - A Saito
- Department of Biochemistry, Tokyo Dental College, Tokyo, Japan.,Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| | - A Saito
- Department of Periodontology, Tokyo Dental College, Tokyo, Japan.,Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| | - T Azuma
- Department of Biochemistry, Tokyo Dental College, Tokyo, Japan.,Oral Health Science Center, Tokyo Dental College, Tokyo, Japan
| |
Collapse
|
29
|
Zheng Z, James AW, Li C, Jiang W, Wang JZ, Chang GX, Lee KS, Chen F, Berthiaume EA, Chen Y, Pan HC, Chen EC, Li W, Zhao Z, Zhang X, Ting K, Soo C. Fibromodulin reduces scar formation in adult cutaneous wounds by eliciting a fetal-like phenotype. Signal Transduct Target Ther 2017; 2:17050-. [PMID: 29201497 PMCID: PMC5661627 DOI: 10.1038/sigtrans.2017.50] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/28/2017] [Accepted: 08/18/2017] [Indexed: 02/02/2023] Open
Abstract
Blocking transforming growth factor (TGF)β1 signal transduction has been a central strategy for scar reduction; however, this approach appears to be minimally effective. Here, we show that fibromodulin (FMOD), a 59-kD small leucine-rich proteoglycan critical for normal collagen fibrillogenesis, significantly reduces scar formation while simultaneously increasing scar strength in both adult rodent models and porcine wounds, which simulate human cutaneous scar repair. Mechanistically, FMOD uncouples pro-migration/contraction cellular signals from pro-fibrotic signaling by selectively enhancing SMAD3-mediated signal transduction, while reducing AP-1-mediated TGFβ1 auto-induction and fibrotic extracellular matrix accumulation. Consequently, FMOD accelerates TGFβ1-responsive adult fibroblast migration, myofibroblast conversion, and function. Furthermore, our findings strongly indicate that, by delicately orchestrating TGFβ1 activities rather than indiscriminately blocking TGFβ1, FMOD elicits fetal-like cellular and molecular phenotypes in adult dermal fibroblasts in vitro and adult cutaneous wounds in vivo, which is a unique response of living system undescribed previously. Taken together, this study illuminates the signal modulating activities of FMOD beyond its structural support functions, and highlights the potential for FMOD-based therapies to be used in cutaneous wound repair.
Collapse
Affiliation(s)
- Zhong Zheng
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Aaron W James
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Chenshuang Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wenlu Jiang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Joyce Z Wang
- Department of Emergency Medicine, Highland General Hospital, Oakland, CA 94602, USA
| | - Grace X Chang
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kevin S Lee
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Feng Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Central Laboratory, School of Stomatology, Peking University, Beijing 100081, China
| | - Emily A Berthiaume
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yao Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hsin Chuan Pan
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Eric C Chen
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Weiming Li
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Orthopaedics, the First Clinical Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, Department of Orthodontics, West China School of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinli Zhang
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Division of Growth and Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, Department of Orthopaedic Surgery, The Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
30
|
Kram V, Kilts TM, Bhattacharyya N, Li L, Young MF. Small leucine rich proteoglycans, a novel link to osteoclastogenesis. Sci Rep 2017; 7:12627. [PMID: 28974711 PMCID: PMC5626712 DOI: 10.1038/s41598-017-12651-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 09/13/2017] [Indexed: 02/01/2023] Open
Abstract
Biglycan (Bgn) and Fibromodulin (Fmod) are subtypes of the small leucine-rich family of proteoglycans (SLRP). In this study we examined the skeletal phenotype of BgnFmod double knockout (BgnFmod KO) mice and found they were smaller in size and have markedly reduced bone mass compared to WT. The low bone mass (LBM) phenotype is the result of both the osteoblasts and osteoclasts from BgnFmod KO mice having higher differentiation potential and being more active compared to WT mice. Using multiple approaches, we showed that both Bgn and Fmod directly bind TNFα as well as RANKL in a dose dependent manner and that despite expressing higher levels of both TNFα and RANKL, BgnFmod KO derived osteoblasts cannot retain these cytokines in the vicinity of the cells, which leads to elevated TNFα and RANKL signaling and enhanced osteoclastogenesis. Furthermore, adding either Bgn or Fmod to osteoclast precursor cultures significantly attenuated the cells ability to form TRAP positive, multinucleated giant cells. In summary, our data indicates that Bgn and Fmod expressed by the bone forming cells, are novel coupling ECM components that control bone mass through sequestration of TNFα and/or RANKL, thereby adjusting their bioavailability in order to regulate osteoclastogenesis.
Collapse
Affiliation(s)
- Vardit Kram
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Tina M Kilts
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nisan Bhattacharyya
- Scientific Review Branch, Division of Extramural Activities, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Li Li
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marian F Young
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
31
|
Yang G, Rothrauff BB, Lin H, Yu S, Tuan RS. Tendon-Derived Extracellular Matrix Enhances Transforming Growth Factor-β3-Induced Tenogenic Differentiation of Human Adipose-Derived Stem Cells. Tissue Eng Part A 2017; 23:166-176. [PMID: 27809678 DOI: 10.1089/ten.tea.2015.0498] [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] [Indexed: 12/19/2022] Open
Abstract
Because of the limited and unsatisfactory outcomes of clinical tendon repair, tissue engineering approaches using adult mesenchymal stem cells are being considered a promising alternative strategy to heal tendon injuries. Successful and functional tendon tissue engineering depends on harnessing the biochemical cues presented by the native tendon extracellular matrix (ECM) and the embedded tissue-specific biofactors. In this study, we have prepared and characterized the biological activities of a soluble extract of decellularized tendon ECM (tECM) on adult adipose-derived stem cells (ASCs), on the basis of histological, biochemical, and gene expression analyses. The results showed that tECM enhances the proliferation and transforming growth factor (TGF)-β3-induced tenogenesis of ASCs in both plate and scaffold cultures in vitro, and modulates matrix deposition of ASCs seeded in scaffolds. These findings suggest that combining tendon ECM extract with TGF-β3 treatment is a possible alternative approach to induce tenogenesis for ASCs.
Collapse
Affiliation(s)
- Guang Yang
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Bioengineering, University of Pittsburgh Swanson School of Engineering , Pittsburgh, Pennsylvania
| | - Benjamin B Rothrauff
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Hang Lin
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| | - Shuting Yu
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,5 School of Medicine, Tsinghua University , Beijing, China
| | - Rocky S Tuan
- 1 Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania.,3 Department of Bioengineering, University of Pittsburgh Swanson School of Engineering , Pittsburgh, Pennsylvania.,4 Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine , Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Ao Z, Yu S, Qian P, Gao W, Guo R, Dong X, Xu J, Zhang R, Jiang C, Ji F, Qian G. Tumor angiogenesis of SCLC inhibited by decreased expression of FMOD via downregulating angiogenic factors of endothelial cells. Biomed Pharmacother 2017; 87:539-547. [PMID: 28081464 DOI: 10.1016/j.biopha.2016.12.110] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 12/14/2016] [Accepted: 12/26/2016] [Indexed: 12/21/2022] Open
Abstract
Fibromodulin (FMOD), an ECM small leucine-rich proteoglycan (SLRP), was reported to promote angiogenesis not only during wound healing, but also in optical and cutaneous angiogenesis-dependent diseases. However, whether it plays important roles in tumor angiogenesis remains unclear. To explore the role of FMOD in tumor angiogenesis of human small cell lung cancer (SCLC), initially the study analyzed the relationship of FMOD expression in cancer tissues of SCLC with clinical characteristics. The analysis revealed that the positive FMOD expression was significantly associated with extensive stage of SCLC and higher vascular density. In mouse models, xenograft tumors developed with FMOD-silenced H446 cells (H446-shFMOD) exhibited slowed growth rate, decreased microvessel density, and reduced blood perfusion related to that of controls (H446-shCON). Additionally, compared with that of controls, the decreased secretion of FMOD in conditioned medium (CM) from H446-shFMOD inhibited proliferation, migration, and invasion of human umbilical vessel endothelial cells (HUVECs). Moreover, the decreased secretion of FMOD downregulated the expression of VEGF, TGF-β1, FGF-2, and PDGF-B in HUVECs. The findings strongly suggested that the autocrine FMOD of cancer cells may promote tumor angiogenesis of SCLC by upregulating the expression of angiogenic factors that act in concert to facilitate the angiogenic phenotype of endothelial cells as a proangiogenic factor. Therefore, silencing FMOD may be a potentially clinical therapy for repressing tumor angiogenesis.
Collapse
Affiliation(s)
- Zhi Ao
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Shilong Yu
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Pin Qian
- Institute of Field Internal Medicine, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Wenhong Gao
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Ruiling Guo
- Department of Respiratory Diseases, 324th Hospital of the People's Liberation Army, Chongqing, 400020, China
| | - Xiaoxiao Dong
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Jianping Xu
- Department of Pathology, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Ruijie Zhang
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Chaowen Jiang
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China
| | - Fuyun Ji
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China.
| | - Guisheng Qian
- Institute of Respiratory Disease, Xinqiao Hospital, The Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
33
|
Jan AT, Lee EJ, Choi I. Fibromodulin: A regulatory molecule maintaining cellular architecture for normal cellular function. Int J Biochem Cell Biol 2016; 80:66-70. [PMID: 27693429 DOI: 10.1016/j.biocel.2016.09.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/23/2016] [Accepted: 09/28/2016] [Indexed: 01/02/2023]
Abstract
Fibromodulin (FMOD) is a small leucine-rich proteoglycan that plays roles in a series of biological and pathophysiological processes. The interaction between FMOD and lysyl oxidase (LOX; collagen cross-linking enzyme) helps regulate extracellular matrix composition, and thereby, provides a permissive environment for regulating cellular turnover. FMOD has been mostly studied in the context of matrix component assembly, but during recent years its association with muscle development, cell reprogramming, and the angiogenic program have demonstrated its activities well beyond extracellular matrix maintenance. In fact, the involvement of FMOD in these cellular processes places it the centrum of cellular behaviour and ultimately of tissue properties. Thus, a clear view of the impact FMOD has on tissue integrity would aid its exploitation for tissue modelling and in the treatment of different disorders.
Collapse
Affiliation(s)
- Arif Tasleem Jan
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
34
|
Zheng Z, Zhang X, Dang C, Beanes S, Chang GX, Chen Y, Li CS, Lee KS, Ting K, Soo C. Fibromodulin Is Essential for Fetal-Type Scarless Cutaneous Wound Healing. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2824-2832. [PMID: 27665369 DOI: 10.1016/j.ajpath.2016.07.023] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 07/11/2016] [Accepted: 07/22/2016] [Indexed: 12/21/2022]
Abstract
In contrast to adult and late-gestation fetal skin wounds, which heal with scar, early-gestation fetal skin wounds display a remarkable capacity to heal scarlessly. Although the underlying mechanism of this transition from fetal-type scarless healing to adult-type healing with scar has been actively investigated for decades, in utero restoration of scarless healing in late-gestation fetal wounds has not been reported. In this study, using loss- and gain-of-function rodent fetal wound models, we identified that fibromodulin (Fm) is essential for fetal-type scarless wound healing. In particular, we found that loss of Fm can eliminate the ability of early-gestation fetal rodents to heal without scar. Meanwhile, administration of fibromodulin protein (FM) alone was capable of restoring scarless healing in late-gestation rat fetal wounds, which naturally heal with scar, as characterized by dermal appendage restoration and organized collagen architectures that were virtually indistinguishable from those in age-matched unwounded skin. High Fm levels correlated with decreased transforming growth factor (TGF)-β1 expression and scarless repair, while low Fm levels correlated with increased TGF-β1 expression and scar formation. This study represents the first successful in utero attempt to induce scarless repair in late-gestation fetal wounds by using a single protein, Fm, and highlights the crucial role that the FM-TGF-β1 nexus plays in fetal-type scarless skin repair.
Collapse
Affiliation(s)
- Zhong Zheng
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, the Department of Orthopaedic Surgery, and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Catherine Dang
- Saul & Joyce Brandman Breast Center, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California
| | | | - Grace X Chang
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Yao Chen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Chen-Shuang Li
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kevin S Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, California; UCLA Division of Plastic and Reconstructive Surgery, the Department of Orthopaedic Surgery, and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California; Department of Bioengineering, School of Engineering, University of California, Los Angeles, Los Angeles, California.
| | - Chia Soo
- UCLA Division of Plastic and Reconstructive Surgery, the Department of Orthopaedic Surgery, and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
35
|
Anderson KJ, Vermillion KL, Jagtap P, Johnson JE, Griffin TJ, Andrews MT. Proteogenomic Analysis of a Hibernating Mammal Indicates Contribution of Skeletal Muscle Physiology to the Hibernation Phenotype. J Proteome Res 2016; 15:1253-61. [DOI: 10.1021/acs.jproteome.5b01138] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- Kyle J. Anderson
- Department
of Biology, University of Minnesota Duluth, 1035 Kirby Drive, Duluth, Minnesota 55812, United States
| | - Katie L. Vermillion
- Department
of Biology, University of Minnesota Duluth, 1035 Kirby Drive, Duluth, Minnesota 55812, United States
| | - Pratik Jagtap
- Center
for Mass Spectrometry and Proteomics, University of Minnesota, 1479 Gortner
Avenue, St. Paul, Minnesota 55108, Unites States
- Department
of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Streey South East, Minneapolis, Minnesota 55455, United States
| | - James E. Johnson
- Minnesota Supercomputing Institute, 512 Walter Library, 117 Pleasant Street South East, Minneapolis, Minnesota 55455, United States
| | - Timothy J. Griffin
- Center
for Mass Spectrometry and Proteomics, University of Minnesota, 1479 Gortner
Avenue, St. Paul, Minnesota 55108, Unites States
- Department
of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, 321 Church Streey South East, Minneapolis, Minnesota 55455, United States
| | - Matthew T. Andrews
- Department
of Biology, University of Minnesota Duluth, 1035 Kirby Drive, Duluth, Minnesota 55812, United States
| |
Collapse
|
36
|
Techniques of Human Embryonic Stem Cell and Induced Pluripotent Stem Cell Derivation. Arch Immunol Ther Exp (Warsz) 2016; 64:349-70. [PMID: 26939778 PMCID: PMC5021740 DOI: 10.1007/s00005-016-0385-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
Abstract
Developing procedures for the derivation of human pluripotent stem cells (PSCs) gave rise to novel pathways into regenerative medicine research. For many years, stem cells have attracted attention as a potentially unlimited cell source for cellular therapy in neurodegenerative disorders, cardiovascular diseases, and spinal cord injuries, for example. In these studies, adult stem cells were insufficient; therefore, many attempts were made to obtain PSCs by other means. This review discusses key issues concerning the techniques of pluripotent cell acquisition. Technical and ethical issues hindered the medical use of somatic cell nuclear transfer and embryonic stem cells. Therefore, induced PSCs (iPSCs) emerged as a powerful technique with great potential for clinical applications, patient-specific disease modelling and pharmaceutical studies. The replacement of viral vectors or the administration of analogous proteins or chemical compounds during cell reprogramming are modifications designed to reduce tumorigenesis risk and to augment the procedure efficiency. Intensified analysis of new PSC lines revealed other barriers to overcome, such as epigenetic memory, disparity between human and mouse pluripotency, and variable response to differentiation of some iPSC lines. Thus, multidimensional verification must be conducted to fulfil strict clinical-grade requirements. Nevertheless, the first clinical trials in patients with spinal cord injury and macular dystrophy were recently carried out with differentiated iPSCs, encouraging alternative strategies for potential autologous cellular therapies.
Collapse
|
37
|
Monaghan MG, Holeiter M, Layland SL, Schenke-Layland K. Cardiomyocyte generation from somatic sources - current status and future directions. Curr Opin Biotechnol 2016; 40:49-55. [PMID: 26945640 DOI: 10.1016/j.copbio.2016.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/11/2016] [Accepted: 02/15/2016] [Indexed: 12/16/2022]
Abstract
Transdifferentiation of one cell type to another has garnered significant research efforts in recent years. As cardiomyocyte loss following myocardial infarction becomes debilitating for cardiac patients, the option of an autologous source of cardiomyocytes not derived from multi/pluripotent stem cell sources is an attractive option. Such direct programming has been clearly realized with the use of transcription factors, microRNAs and more recently small molecule delivery to enhance epigenetic modifications, all albeit with low efficiencies in vitro. In this review, we aim to present a brief overview of the current in vitro and in vivo transdifferentiation strategies in the generation of cardiomyocytes from somatic sources. The interdisciplinary fields of tissue, cell, material and regenerative engineering offer many opportunities to synergistically achieve directly programmed cardiac tissue in vitro and enhance transdifferentiation in vivo. This review aims to present a concise outlook on this topic with these fields in mind.
Collapse
Affiliation(s)
- Michael G Monaghan
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Monika Holeiter
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany
| | - Shannon L Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany; Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB), Stuttgart, Germany
| | - Katja Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, Eberhard Karls University, Tübingen, Germany; Department of Cell and Tissue Engineering, Fraunhofer Institute for Interfacial Engineering and Biotechnology (IGB), Stuttgart, Germany; Department of Medicine/Cardiology, Cardiovascular Research Laboratories, University of California, Los Angeles, CA, USA.
| |
Collapse
|
38
|
Li CS, Yang P, Ting K, Aghaloo T, Lee S, Zhang Y, Khalilinejad K, Murphy MC, Pan HC, Zhang X, Wu B, Zhou YH, Zhao Z, Zheng Z, Soo C. Fibromodulin reprogrammed cells: A novel cell source for bone regeneration. Biomaterials 2016; 83:194-206. [PMID: 26774565 PMCID: PMC4754141 DOI: 10.1016/j.biomaterials.2016.01.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 12/27/2015] [Accepted: 01/01/2016] [Indexed: 02/05/2023]
Abstract
Pluripotent or multipotent cell-based therapeutics are vital for skeletal reconstruction in non-healing critical-sized defects since the local endogenous progenitor cells are not often adequate to restore tissue continuity or function. However, currently available cell-based regenerative strategies are hindered by numerous obstacles including inadequate cell availability, painful and invasive cell-harvesting procedures, and tumorigenesis. Previously, we established a novel platform technology for inducing a quiescent stem cell-like stage using only a single extracellular proteoglycan, fibromodulin (FMOD), circumventing gene transduction. In this study, we further purified and significantly increased the reprogramming rate of the yield multipotent FMOD reprogrammed (FReP) cells. We also exposed the 'molecular blueprint' of FReP cell osteogenic differentiation by gene profiling. Radiographic analysis showed that implantation of FReP cells into a critical-sized SCID mouse calvarial defect, contributed to the robust osteogenic capability of FReP cells in a challenging clinically relevant traumatic scenario in vivo. The persistence, engraftment, and osteogenesis of transplanted FReP cells without tumorigenesis in vivo were confirmed by histological and immunohistochemical staining. Taken together, we have provided an extended potency, safety, and molecular profile of FReP cell-based bone regeneration. Therefore, FReP cells present a high potential for cellular and gene therapy products for bone regeneration.
Collapse
Affiliation(s)
- Chen-Shuang Li
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Pu Yang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Kang Ting
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Tara Aghaloo
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Soonchul Lee
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA; Department of Orthopaedic Surgery, CHA Bundang Medical Center, CHA University, Gyeonggi-do, 463-712, South Korea
| | - Yulong Zhang
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kambiz Khalilinejad
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Maxwell C Murphy
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin Chuan Pan
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin Wu
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yan-Heng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, PR China
| | - Zhong Zheng
- Dental and Craniofacial Research Institute and Division of Growth and Development, Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Chia Soo
- UCLA Division of Plastic Surgery and Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
39
|
Hultgårdh-Nilsson A, Borén J, Chakravarti S. The small leucine-rich repeat proteoglycans in tissue repair and atherosclerosis. J Intern Med 2015; 278:447-61. [PMID: 26477596 PMCID: PMC4616156 DOI: 10.1111/joim.12400] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteoglycans consist of a protein core with one or more covalently attached glycosaminoglycan (GAG) side chains and have multiple roles in the initiation and progression of atherosclerosis. Here we discuss the potential and known functions of a group of small leucine-rich repeat proteoglycans (SLRPs) in atherosclerosis. We focus on five SLRPs, decorin, biglycan, lumican, fibromodulin and PRELP, because these have been detected in atherosclerotic plaques or demonstrated to have a role in animal models of atherosclerosis. Decorin and biglycan are modified post-translationally by substitution with chondroitin/dermatan sulphate GAGs, whereas lumican, fibromodulin and PRELP have keratan sulphate side chains, and the core proteins have leucine-rich repeat (LRR) motifs that are characteristic of the LRR superfamily. The chondroitin/dermatan sulphate GAG side chains have been implicated in lipid retention in atherosclerosis. The core proteins are discussed here in the context of (i) interactions with collagens and their implications in tissue integrity, fibrosis and wound repair and (ii) interactions with growth factors, cytokines, pathogen-associated molecular patterns and cell surface receptors that impact normal physiology and disease processes such as inflammation, innate immune responses and wound healing (i.e. processes that are all important in plaque development and progression). Thus, studies of these SLRPs in the context of wound healing are providing clues about their functions in early stages of atherosclerosis to plaque vulnerability and cardiovascular disease at later stages. Understanding of signal transduction pathways regulated by the core protein interactions is leading to novel roles and therapeutic potential for these proteins in wound repair and atherosclerosis.
Collapse
Affiliation(s)
| | - J Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - S Chakravarti
- Departments of Medicine, Ophthalmology and Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
40
|
Chen L, Liao J, Klineberg E, Leung VYL, Huang S. Small leucine-rich proteoglycans (SLRPs): characteristics and function in the intervertebral disc. J Tissue Eng Regen Med 2015; 11:602-608. [PMID: 26370612 DOI: 10.1002/term.2067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Revised: 04/16/2015] [Accepted: 06/12/2015] [Indexed: 02/05/2023]
Affiliation(s)
- Lili Chen
- Research Centre for Human Tissues and Organs Degeneration; Shenzhen Institute of Advanced Technology, Chinese Academy of Science; Shenzhen 518055 China
| | - Jingwen Liao
- School of Materials Science and Engineering; South China University of Technology; Guangzhou China
| | - Eric Klineberg
- Department of Orthopaedics; University of California at Davis; Sacramento California USA
| | - Victor YL Leung
- Department of Orthopaedics and Traumatology; Li Ka Shing Faculty of Medicine, The University of Hong Kong; Hong Kong
| | - Shishu Huang
- Department of Orthopaedic Surgery; West China Hospital; State Key Laboratory of Oral Diseases, Sichuan University; Chengdu China
- Research Centre for Human Tissues and Organs Degeneration; Shenzhen Institute of Advanced Technology, Chinese Academy of Science; Shenzhen 518055 China
| |
Collapse
|
41
|
Mirakhori F, Zeynali B, Kiani S, Baharvand H. Brief azacytidine step allows the conversion of suspension human fibroblasts into neural progenitor-like cells. CELL JOURNAL 2015; 17:153-8. [PMID: 25870845 PMCID: PMC4393664 DOI: 10.22074/cellj.2015.522] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Accepted: 03/10/2014] [Indexed: 12/29/2022]
Abstract
In recent years transdifferentiation technology has enabled direct conversion of human
fibroblasts to become a valuable, abundant and accessible cell source for patient-specific
induced cell generation in biomedical research. The majority of transdifferentiation approaches rely upon viral gene delivery which due to random integration with the host
genome can cause genome instability and tumorigenesis upon transplantation. Here, we
provide a simple way to induce neural progenitor-like cells from human fibroblasts without genetic manipulation by changing physicochemical culture properties from monolayer
culture into a suspension in the presence of a chemical DNA methyltransferase inhibitor
agent, Azacytidine. We have demonstrated the expression of neural progenitor-like markers, morphology and the ability to spontaneously differentiate into neural-like cells. This
approach is simple, inexpensive, lacks genetic manipulation and could be a foundation for
future chemical neural transdifferentiation and a safe induction of neural progenitor cells
from human fibroblasts for clinical applications.
Collapse
Affiliation(s)
- Fahimeh Mirakhori
- School of Biology, College of Science, University of Tehran, Tehran, Iran ; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Bahman Zeynali
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Sahar Kiani
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran ; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| |
Collapse
|
42
|
Fibromodulin Enhances Angiogenesis during Cutaneous Wound Healing. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2015; 2:e275. [PMID: 25587509 PMCID: PMC4292257 DOI: 10.1097/gox.0000000000000243] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/06/2014] [Indexed: 12/20/2022]
Abstract
Supplemental Digital Content is available in the text. Background: Fibromodulin (FMOD) plays a critical role in the wound-healing process. Our previous studies revealed that FMOD deficiency led to marked alterations in adult wound healing characterized by delayed dermal cell migration, postponed wound closure, and increased scar formation, all accompanied by impeded angiogenesis. Therefore, the aim of this study was to reveal the effect of FMOD on angiogenesis during the wound-healing process. Methods: In vivo angiogenic effects of FMOD were assessed by a chick embryo chorioallantoic membrane assay, a Matrigel (BD Bioscience, Franklin Lakes, N.J.) plug implant assay, and rodent primary closure wound models. In vitro angiogenic effects of FMOD were recorded by cell invasion and dimensional and topological parameters of human umbilical vein endothelial cells. Results: We provided evidence that FMOD significantly enhanced vascularization: first, FMOD boosted blood vessel formation on the chorioallantoic membrane; second, FMOD markedly stimulated capillary infiltration into Matrigel plugs subcutaneously implanted in adult mice; and finally, FMOD robustly promoted angiogenesis in multiple adult rodent cutaneous wound models. Furthermore, FMOD administration restored the vascularity of fmod−/− mouse wounds. In support of this, FMOD endorsed an angiogenesis-favored microenvironment in adult rodent wounds not only by upregulating angiogenic genes but also by downregulating angiostatic genes. In addition, FMOD significantly enhanced human umbilical vein endothelial cell invasion and tube-like structure formation in vitro. Conclusions: Altogether, we demonstrated that in addition to reducing scar formation, FMOD also promotes angiogenesis. As blood vessels organize and regulate wound healing, its potent angiogenic properties will further expand the clinical application of FMOD for cutaneous healing of poorly vascularized wounds.
Collapse
|
43
|
Generation of pluripotent stem cells without the use of genetic material. J Transl Med 2015; 95:26-42. [PMID: 25365202 DOI: 10.1038/labinvest.2014.132] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/25/2014] [Accepted: 07/25/2014] [Indexed: 01/18/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) provide a platform to obtain patient-specific cells for use as a cell source in regenerative medicine. Although iPSCs do not have the ethical concerns of embryonic stem cells, iPSCs have not been widely used in clinical applications, as they are generated by gene transduction. Recently, iPSCs have been generated without the use of genetic material. For example, protein-induced PSCs and chemically induced PSCs have been generated by the use of small and large (protein) molecules. Several epigenetic characteristics are important for cell differentiation; therefore, several small-molecule inhibitors of epigenetic-modifying enzymes, such as DNA methyltransferases, histone deacetylases, histone methyltransferases, and histone demethylases, are potential candidates for the reprogramming of somatic cells into iPSCs. In this review, we discuss what types of small chemical or large (protein) molecules could be used to replace the viral transduction of genes and/or genetic reprogramming to obtain human iPSCs.
Collapse
|
44
|
Wang PY, Lee HHC, Higuchi A, Ling QD, Lin HR, Li HF, Suresh Kumar S, Chang Y, Alarfaj AA, Munusamy MA, Chen DC, Hsu ST, Wang HC, Hsiao HY, Wu GJ. Pluripotency maintenance of amniotic fluid-derived stem cells cultured on biomaterials. J Mater Chem B 2015; 3:3858-3869. [DOI: 10.1039/c5tb00447k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Human amniotic fluid-derived stem cells can maintain their pluripotency when cultured on soft hydrogels.
Collapse
|
45
|
Zheng Z, Lee KS, Zhang X, Nguyen C, Hsu C, Wang JZ, Rackohn TM, Enjamuri DR, Murphy M, Ting K, Soo C. Fibromodulin-deficiency alters temporospatial expression patterns of transforming growth factor-β ligands and receptors during adult mouse skin wound healing. PLoS One 2014; 9:e90817. [PMID: 24603701 PMCID: PMC3948369 DOI: 10.1371/journal.pone.0090817] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Accepted: 02/04/2014] [Indexed: 12/17/2022] Open
Abstract
Fibromodulin (FMOD) is a small leucine-rich proteoglycan required for scarless fetal cutaneous wound repair. Interestingly, increased FMOD levels have been correlated with decreased transforming growth factor (TGF)-β1 expression in multiple fetal and adult rodent models. Our previous studies demonstrated that FMOD-deficiency in adult animals results in delayed wound closure and increased scar size accompanied by loose package collagen fiber networks with increased fibril diameter. In addition, we found that FMOD modulates in vitro expression and activities of TGF-β ligands in an isoform-specific manner. In this study, temporospatial expression profiles of TGF-β ligands and receptors in FMOD-null and wild-type (WT) mice were compared by immunohistochemical staining and quantitative reverse transcriptase-polymerase chain reaction using a full-thickness, primary intention wound closure model. During the inflammatory stage, elevated inflammatory infiltration accompanied by increased type I TGF-β receptor levels in individual inflammatory cells was observed in FMOD-null wounds. This increased inflammation was correlated with accelerated epithelial migration during the proliferative stage. On the other hand, significantly more robust expression of TGF-β3 and TGF-β receptors in FMOD-null wounds during the proliferative stage was associated with delayed dermal cell migration and proliferation, which led to postponed granulation tissue formation and wound closure and increased scar size. Compared with WT controls, expression of TGF-β ligands and receptors by FMOD-null dermal cells was markedly reduced during the remodeling stage, which may have contributed to the declined collagen synthesis capability and unordinary collagen architecture. Taken together, this study demonstrates that a single missing gene, FMOD, leads to conspicuous alternations in TGF-β ligand and receptor expression at all stages of wound repair in various cell types. Therefore, FMOD critically coordinates temporospatial distribution of TGF-β ligands and receptors in vivo, suggesting that FMOD modulates TGF-β bioactivity in a complex way beyond simple physical binding to promote proper wound healing.
Collapse
Affiliation(s)
- Zhong Zheng
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kevin S. Lee
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Calvin Nguyen
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Chingyun Hsu
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Joyce Z. Wang
- Department of Emergency Medicine, State University of New York Downstate/Kings Country Hospital Center, New York, New York, United States of America
| | - Todd Matthew Rackohn
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Dwarak Reddy Enjamuri
- Department of Psychobiology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Maxwell Murphy
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California Los Angeles, Los Angeles, California, United States of America
| | - Chia Soo
- UCLA and Orthopaedic Hospital Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, University of California Los Angeles, Los Angeles, California, United States of America
- Division of Plastic and Reconstructive Surgery, Department of Surgery, School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
46
|
Jeon SY, Park JS, Yang HN, Woo DG, Park KH. Aggrecan- and COMP-loaded poly-(lactic-co-glycolic acid) nanoparticles stimulate chondrogenic differentiation of human mesenchymal stem cells. Stem Cells Dev 2013; 23:305-17. [PMID: 24028375 DOI: 10.1089/scd.2013.0311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
During embryogenesis, specific proteins expressed in cells have key roles in the formation of differentiated cells and tissues. Delivery of specific proteins into specific cells, both in vitro and in vivo, has proved to be exceedingly difficult. In this study, we developed a safe and efficient protein delivery system using encapsulation of proteins into biodegradable poly-(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs). The PLGA NPs were used to deliver proteins into human mesenchymal stem cells (hMSCs). Fluorescent markers loaded into the PLGA NPs were used to verify the internalization of NPs into hMSCs using FACS analysis and confocal microscopy. With these methods, we demonstrated that the encapsulated model proteins are readily delivered into hMSCs, released from the NP vehicles, and, finally, moved into the cytosols. Using chondrogenesis-related proteins such as aggrecan and cartilage oligomeric matrix protein (COMP), chondrogenic differentiation of hMSCs treated with aggrecan and COMP encapsulated PLGA NPs was clearly observed and caused to differentiate into chondrocytes.
Collapse
Affiliation(s)
- Su Yeon Jeon
- 1 Department of Biomedical Science, College of Life Science, CHA University , Seongnam-si, Republic of Korea
| | | | | | | | | |
Collapse
|
47
|
Fournier BPJ, Larjava H, Häkkinen L. Gingiva as a source of stem cells with therapeutic potential. Stem Cells Dev 2013; 22:3157-77. [PMID: 23944935 DOI: 10.1089/scd.2013.0015] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Postnatal connective tissues contain phenotypically heterogeneous cells populations that include distinct fibroblast subpopulations, pericytes, myofibroblasts, fibrocytes, and tissue-specific mesenchymal stem cells (MSCs). These cells play key roles in tissue development, maintenance, and repair and contribute to various pathologies. Depending on the origin of tissue, connective tissue cells, including MSCs, have different phenotypes. Understanding the identity and specific functions of these distinct tissue-specific cell populations may allow researchers to develop better treatment modalities for tissue regeneration and find novel approaches to prevent pathological conditions. Interestingly, MSCs from adult oral mucosal gingiva possess distinct characteristics, including neural crest origin, multipotent differentiation capacity, fetal-like phenotype, and potent immunomodulatory properties. These characteristics and an easy, relatively noninvasive access to gingival tissue, and fast tissue regeneration after tissue biopsy make gingiva an attractive target for cell isolation for therapeutic purposes aiming to promote tissue regeneration and fast, scar-free wound healing. The purpose of this review is to discuss the identity, phenotypical heterogeneity, and function of gingival MSCs and summarize what is currently known about their properties, role in scar-free healing, and their future therapeutic potential.
Collapse
Affiliation(s)
- Benjamin P J Fournier
- 1 Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia , Vancouver, Canada
| | | | | |
Collapse
|
48
|
Yang G, Rothrauff BB, Lin H, Gottardi R, Alexander PG, Tuan RS. Enhancement of tenogenic differentiation of human adipose stem cells by tendon-derived extracellular matrix. Biomaterials 2013; 34:9295-306. [PMID: 24044998 DOI: 10.1016/j.biomaterials.2013.08.054] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 08/19/2013] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cells (MSCs) have gained increasing research interest for their potential in improving healing and regeneration of injured tendon tissues. Developing functional three-dimensional (3D) scaffolds to promote MSC proliferation and differentiation is a critical requirement in tendon tissue engineering. Tendon extracellular matrix has been shown to maintain the tenogenic potential of tendon stem cells and stimulate tenogenesis of human adipose stem cells (hASCs) in 2D culture. This study aims at characterizing the biological composition of urea-extracted fraction of tendon ECM (tECM) and its tenogenic effect on hASCs cultured in a 3D collagen scaffold under uniaxial tension. The tECM obtained was cell-free and rich in ECM proteins. hASCs seeded in tECM-supplemented scaffold exhibited significantly increased proliferation and tenogenic differentiation. The presence of tECM also greatly suppressed the osteogenic differentiation of hASCs triggered by uniaxial tension. In addition, tECM-supplemented constructs displayed enhanced mechanical strength, accompanied by reduced expression and activity of MMPs in the seeded hASCs, indicating a regulatory activity of tECM in cell-mediated scaffold remodeling. These findings support the utility of tECM in creating bio-functional scaffolds for tendon tissue engineering.
Collapse
Affiliation(s)
- Guang Yang
- Center for Cellular and Molecular Engineering, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15219, USA
| | | | | | | | | | | |
Collapse
|
49
|
Jian J, Zheng Z, Zhang K, Rackohn TM, Hsu C, Levin A, Enjamuri DR, Zhang X, Ting K, Soo C. Fibromodulin promoted in vitro and in vivo angiogenesis. Biochem Biophys Res Commun 2013; 436:530-535. [PMID: 23770359 DOI: 10.1016/j.bbrc.2013.06.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 06/03/2013] [Indexed: 12/20/2022]
Abstract
Fibromodulin (FMOD) is an extracellular matrix (ECM) small leucine-rich proteoglycan (SLRP) that plays an important role in cell fate determination. Previous studies revealed that not only is FMOD critical in fetal-type scarless wound healing, but it also promotes adult wound closure and reduces scar formation. In addition, FMOD-deficient mice exhibit significantly reduced blood vessel regeneration in granulation tissues during wound healing. In this study, we investigated the effects of FMOD on angiogenesis, which is an important event in wound healing as well as embryonic development and tumorigenesis. We found that FMOD accelerated human umbilical vein endothelial HUVEC-CS cell adhesion, spreading, actin stress fiber formation, and eventually tube-like structure (TLS) network establishment in vitro. On a molecular level, by increasing expression of collagen I and III, angiopoietin (Ang)-2, and vascular endothelial growth factor (VEGF), as well as reducing the ratio of Ang-1/Ang-2, FMOD provided a favorable network to mobilize quiescent endothelial cells to an angiogenic phenotype. Moreover, we also confirmed that FMOD enhanced angiogenesis in vivo by using an in ovo chick embryo chorioallantoic membrane (CAM) assay. Therefore, our data demonstrate that FMOD is a pro-angiogenic and suggest a potential therapeutic role of FMOD in the treatment of conditions related to impaired angiogenesis.
Collapse
Affiliation(s)
- Jia Jian
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zhong Zheng
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kermit Zhang
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Todd Matthew Rackohn
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chingyun Hsu
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew Levin
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Dwarak Reddy Enjamuri
- Department of Psychobiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xinli Zhang
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kang Ting
- Dental and Craniofacial Research Institute and Section of Orthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery and Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
50
|
Tavernier G, Mlody B, Demeester J, Adjaye J, De Smedt SC. Current methods for inducing pluripotency in somatic cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:2765-2771. [PMID: 23529911 DOI: 10.1002/adma.201204874] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Indexed: 06/02/2023]
Abstract
The groundbreaking discovery of reprogramming fibroblasts towards pluripotency merely by introducing four transcription factors (OCT4, SOX2, KLF4 and c-MYC) by means of retroviral transduction has created a promising revolution in the field of regenerative medicine. These so-called induced pluripotent stem cells (iPSCs) can provide a cell source for disease-modelling, drug-screening platforms, and transplantation strategies to treat incurable degenerative diseases, while circumventing the ethical issues and immune rejections associated with the use of non-autologous embryonic stem cells. The risk of insertional mutagenesis, caused both by the viral and transgene nature of the technique has proven to be the major limitation for iPSCs to be used in a clinical setting. In view of this, a variety of alternative techniques have been developed to induce pluripotency in somatic cells. This review provides an overview on current reprogramming protocols, discusses their pros and cons and future challenges to provide safe and transgene-free iPSCs.
Collapse
Affiliation(s)
- Geertrui Tavernier
- Ghent University, Laboratory of General Biochemistry and Physical Pharmacy, Ghent Research Group on Nanomedicines, Harelbekestraat 72, Ghent, Belgium
| | | | | | | | | |
Collapse
|