1
|
Tsuchiya M, Ohashi Y, Kodera Y, Satoh M, Matsui T, Fukushima K, Iwase D, Aikawa J, Mukai M, Inoue G, Takaso M, Uchida K. CD39+CD55- Fb Subset Exhibits Myofibroblast-Like Phenotype and Is Associated with Pain in Osteoarthritis of the Knee. Biomedicines 2023; 11:3047. [PMID: 38002046 PMCID: PMC10669511 DOI: 10.3390/biomedicines11113047] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Recent studies utilizing single-cell analysis have unveiled the presence of various fibroblast (Fb) subsets within the synovium under inflammatory conditions in osteoarthritis (OA), distinguishing them from those in rheumatoid arthritis (RA). Moreover, it has been reported that pain in knee OA patients is linked to specific fibroblast subsets. Single-cell expression profiling methods offer an incredibly detailed view of the molecular states of individual cells. However, one limitation of these methods is that they require the destruction of cells during the analysis process, rendering it impossible to directly assess cell function. In our study, we employ flow cytometric analysis, utilizing cell surface markers CD39 and CD55, in an attempt to isolate fibroblast subsets and investigate their relationship with OA pathology. Synovial tissues were obtained from 25 knee OA (KOA) patients. Of these, six samples were analyzed by RNA-seq (n = 3) and LC/MS analysis (n = 3). All 25 samples were analyzed to estimate the proportion of Fb (CD45-CD31-CD90+) subset by flow cytometry. The proportion of Fb subsets (CD39+CD55- and CD39-CD55+) and their association with osteoarthritis pathology were evaluated. CD39+CD55- Fb highly expressed myogenic markers such as CNN1, IGFBP7, MYH11, and TPM1 compared to CD39-CD55+ Fb. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis of upregulated differentially expressed genes (DEGs) in CD39+CD55- Fb identified the Apelin pathway and cGMP-PKC-signaling pathway as possibly contributing to pain. LC/MS analysis indicated that proteins encoded by myogenic marker genes, including CNN1, IGFBP7, and MYH11, were also significantly higher than in CD39-CD55+ Fb. CD39-CD55+ Fb highly expressed PRG4 genes and proteins. Upregulated DEGs were enriched for pathways associated with proinflammatory states ('RA', 'TNF signaling pathway', 'IL-17 signaling pathway'). The proportion of CD39+CD55- Fb in synovium significantly correlated with both resting and active pain levels in knee OA (KOA) patients (resting pain, ρ = 0.513, p = 0.009; active pain, ρ = 0.483, p = 0.015). There was no correlation between joint space width (JSW) and the proportion of CD39+CD55- Fb. In contrast, there was no correlation between the proportion of CD39-CD55+ Fb and resting pain, active pain, or JSW. In conclusion, CD39+CD55- cells exhibit a myofibroblast phenotype, and its proportion is associated with KOA pain. Our study sheds light on the potential significance of CD39+CD55- synovial fibroblasts in osteoarthritis, their myofibroblast-like phenotype, and their association with joint pain. These findings provide a foundation for further research into the mechanisms underlying fibrosis, the impact of altered gene expression on osteoarthritic joints, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Maho Tsuchiya
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Yoshihisa Ohashi
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Yoshio Kodera
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan; (Y.K.); (T.M.)
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan
| | - Masashi Satoh
- Department of Immunology, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan;
| | - Takashi Matsui
- Department of Physics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan; (Y.K.); (T.M.)
- Center for Disease Proteomics, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara 252-0373, Kanagawa, Japan
| | - Kensuke Fukushima
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Dai Iwase
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Jun Aikawa
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Manabu Mukai
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Gen Inoue
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Masashi Takaso
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| | - Kentaro Uchida
- Department of Orthopaedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku, Kitasato, Sagamihara 252-0374, Kanagawa, Japan; (M.T.); (Y.O.); (K.F.); (D.I.); (J.A.); (M.M.); (G.I.); (M.T.)
| |
Collapse
|
2
|
MiR-340 Promotes the Proliferation of Vascular Smooth Muscle Cells by Targeting von Hippel-Lindau Tumor Suppressor Gene. J Cardiovasc Pharmacol 2021; 77:875-884. [PMID: 34016842 DOI: 10.1097/fjc.0000000000001016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/20/2021] [Indexed: 11/27/2022]
Abstract
ABSTRACT MiRNAs play key roles in the proliferation of vascular smooth muscle cells (VSMCs). However, the roles and underlying mechanism of miRNAs in VSMCs are not fully understood. The aim of this study was to evaluate the role of miR-340 in the proliferation of VSMCs. The expression levels of miR-340 and von Hippel-Lindau tumor suppressor (VHL) in VSMCs induced by platelet-derived growth factor-BB or fetal bovine serum were measured by q-polymerase chain reaction. The effects of miR-340 and VHL on cell proliferation and invasion were evaluated by CCK-8 assay. Target gene prediction and screening as well as luciferase reporter assay were performed to verify the downstream target genes of miR-340. Western blotting was used to detect the protein expression levels of vascular endothelial growth factor and VHL. Our results showed that the miR-340 was upregulated in platelet-derived growth factor-BBor fetal bovine serum-induced VSMCs. In addition, overexpression of miR-340 promoted VSMCs proliferation and invasion. Moreover, VHL was found to be a potential target for miR-340 and upregulation of VHL-inhibited VSMCs proliferation. MiR-340 plays a critical role in VSMC proliferation and neointimal hyperplasia in rats' carotid balloon injury model. Reduced expression levels of miR-340 promoted VHL-inhibited VSMCs proliferation. In conclusion, miR-340 may play a role in the regulation of proliferation of VSMCs by inhibition of VHL.
Collapse
|
3
|
Geelhoed WJ, van der Bogt KEA, Rothuizen TC, Damanik FFR, Hamming JF, Mota CD, van Agen MS, de Boer HC, Restrepo MT, Hinz B, Kislaya A, Poelma C, van Zonneveld AJ, Rabelink TJ, Moroni L, Rotmans JI. A novel method for engineering autologous non-thrombogenic in situ tissue-engineered blood vessels for arteriovenous grafting. Biomaterials 2019; 229:119577. [PMID: 31704466 DOI: 10.1016/j.biomaterials.2019.119577] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 10/14/2019] [Accepted: 10/22/2019] [Indexed: 12/15/2022]
Abstract
The durability of prosthetic arteriovenous (AV) grafts for hemodialysis access is low, predominantly due to stenotic lesions in the venous outflow tract and infectious complications. Tissue engineered blood vessels (TEBVs) might offer a tailor-made autologous alternative for prosthetic grafts. We have designed a method in which TEBVs are grown in vivo, by utilizing the foreign body response to subcutaneously implanted polymeric rods in goats, resulting in the formation of an autologous fibrocellular tissue capsule (TC). One month after implantation, the polymeric rod is extracted, whereupon TCs (length 6 cm, diameter 6.8 mm) were grafted as arteriovenous conduit between the carotid artery and jugular vein of the same goats. At time of grafting, the TCs were shown to have sufficient mechanical strength in terms of bursting pressure (2382 ± 129 mmHg), and suture retention strength (SRS: 1.97 ± 0.49 N). The AV grafts were harvested at 1 or 2 months after grafting. In an ex vivo whole blood perfusion system, the lumen of the vascular grafts was shown to be less thrombogenic compared to the initial TCs and ePTFE grafts. At 8 weeks after grafting, the entire graft was covered with an endothelial layer and abundant elastin expression was present throughout the graft. Patency at 1 and 2 months was comparable with ePTFE AV-grafts. In conclusion, we demonstrate the remodeling capacity of cellularized in vivo engineered TEBVs, and their potential as autologous alternative for prosthetic vascular grafts.
Collapse
Affiliation(s)
- W J Geelhoed
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, the Netherlands
| | - K E A van der Bogt
- Department of Surgery, Leiden University Medical Center, the Netherlands
| | - T C Rothuizen
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands
| | - F F R Damanik
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - J F Hamming
- Department of Surgery, Leiden University Medical Center, the Netherlands
| | - C D Mota
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - M S van Agen
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, the Netherlands
| | - H C de Boer
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, the Netherlands
| | - M Tobón Restrepo
- Division of Diagnostic Imaging, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - B Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Canada
| | - A Kislaya
- Laboratory for Aero and Hydrodynamics, Delft University of Technology, Delft, the Netherlands
| | - C Poelma
- Laboratory for Aero and Hydrodynamics, Delft University of Technology, Delft, the Netherlands
| | - A J van Zonneveld
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory of Vascular and Regenerative Medicine, the Netherlands
| | - T J Rabelink
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands
| | - L Moroni
- MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - J I Rotmans
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands.
| |
Collapse
|
4
|
Nguyen MTN, Tran HLB. Effect of Modified Bovine Pericardium on Human Gingival Fibroblasts in vitro. Cells Tissues Organs 2019; 206:296-307. [PMID: 31357195 DOI: 10.1159/000501807] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022] Open
Abstract
Supportive membranes have recently been applied to treat periodontal disease in order to achieve periodontal tissue regeneration. The crucial role of these membranes is to facilitate the restoration of the structural and functional periodontium. Bovine pericardium (BP) is mainly composed of collagen type I, which was demonstrated to have good mechanical properties and biological regenerative potential. Our research aimed to extend the application of membrane derived from BP to periodontal disease treatment. However, the fabrication method to achieve a xenogenic-free membrane with the mechanical properties required for periodontal treatment is rarely mentioned. Therefore, a procedure for the extraction and modification of BP using sodium dodecyl sulfate (SDS) and glutaraldehyde (GA) was developed. BP was harvested and decellularized using different SDS concentrations (0.05-0.3%). GA was used to further modify the membranes to achieve suitable thickness, mechanical strength, and pore size. A combination protocol of 0.15% SDS treatment for 12 h with continuous agitation combined with 0.1% GA for 6 h for membrane fabricating was applied. The modified BP (mBP) had the targeted characteristics, such as 0.2-0.5 mm thickness, approximately 10 MPa in tensile strength, 30% in strain force, and a pore size <5 µm, which is comparable to commercially available collagen membranes. Findings from this study demonstrated that the established method was effective in preparing BP membrane for periodontal treatment while decreasing the concentration of reagents and processing time. Moreover, our modified membrane was found to have no cytotoxicity but supports the migration, attachment, and proliferation of human gingival fibroblasts in vitro. Taken together, these results confirmed that mBP is suitable for application in periodontal disease treatment and regeneration.
Collapse
Affiliation(s)
- My Thi Ngoc Nguyen
- Laboratory of Tissue Engineering and Biomedical Materials, Department of Physiology and Animal Biotechnology, University of Science, Vietnam National University, Ho Chi Minh, Vietnam
| | - Ha Le Bao Tran
- Laboratory of Tissue Engineering and Biomedical Materials, Department of Physiology and Animal Biotechnology, University of Science, Vietnam National University, Ho Chi Minh, Vietnam,
| |
Collapse
|
5
|
Chen X, Chen Y, Hou Y, Song P, Zhou M, Nie M, Liu X. Modulation of proliferation and differentiation of gingiva‑derived mesenchymal stem cells by concentrated growth factors: Potential implications in tissue engineering for dental regeneration and repair. Int J Mol Med 2019; 44:37-46. [PMID: 31017269 PMCID: PMC6559294 DOI: 10.3892/ijmm.2019.4172] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/10/2019] [Indexed: 12/16/2022] Open
Abstract
The aim of the present study was to evaluate the proliferation and osteogenic differentiation ability of gingiva-derived mesenchymal stem cells (GMSCs) cultured with different concentrations of concentrated growth factors (CGF). GMSCs were isolated from gingival connective tissues and characterized by flow cytometry, immunofluorescence staining and immunohistochemical staining. Cell proliferation activity was determined by the MTT assay, and the effect of CGF on MCSCs was detected with the Cell Counting Kit (CCK)-8 assay. Mineralization induction was evaluated by alkaline phosphatase (ALP)-positive cell staining and mineralized nodule formation assay. Dentin matrix acidic phosphoprotein (DMP)1, dentin sialophosphoprotein (DSPP), bone morphogenetic protein (BMP)2 and runt-related transcription factor (RUNX)2 mRNA and protein expression were evaluated by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis and western blotting. The flow cytometry, immunofluorescence staining and immunohistochemical staining results indicated that the cultured cells were GMSCs. The MTT assay results revealed that the third-generation gingival stem cells exhibited the highest proliferative capacity, and the CCK-8 results indicated that 10% CGF achieved the most prominent promotion of GMSC proliferation. ALP activity analysis and mineralized nodule assay demonstrated that CGF may successfully induce osteogenic differentiation of GMSCs, whereas RT-qPCR and western blot analyses demonstrated that CGF is involved in the differentiation of GMSCs by regulating the expression of DMP1, DSPP, BMP2 and RUNX2 (P<0.05). In conclusion, CGF were demonstrated to promote the proliferation and osteogenic differentiation of GMSCs. Therefore, CGF may be applied in tissue engineering for tooth regeneration and repair.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Orthodontics, Mianyang Stomatological Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Yuhe Chen
- Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Yali Hou
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Peng Song
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Minyue Zhou
- Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Minhai Nie
- Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xuqian Liu
- Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
6
|
Qiu YL, Chen X, Hou YL, Hou YJ, Tian SB, Chen YH, Yu L, Nie MH, Liu XQ. Characterization of different biodegradable scaffolds in tissue engineering. Mol Med Rep 2019; 19:4043-4056. [PMID: 30896809 PMCID: PMC6471812 DOI: 10.3892/mmr.2019.10066] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 12/19/2018] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to compare the characteristics of acellular dermal matrix (ADM), small intestinal submucosa (SIS) and Bio‑Gide scaffolds with acellular vascular matrix (ACVM)‑0.25% human‑like collagen I (HLC‑I) scaffold in tissue engineering blood vessels. The ACVM‑0.25% HLC‑I scaffold was prepared and compared with ADM, SIS and Bio‑Gide scaffolds via hematoxylin and eosin (H&E) staining, Masson staining and scanning electron microscope (SEM) observations. Primary human gingival fibroblasts (HGFs) were cultured and identified. Then, the experiment was established via the seeding of HGFs on different scaffolds for 1, 4 and 7 days. The compatibility of four different scaffolds with HGFs was evaluated by H&E staining, SEM observation and Cell Counting Kit‑8 assay. Then, a series of experiments were conducted to evaluate water absorption capacities, mechanical abilities, the ultra‑microstructure and the cytotoxicity of the four scaffolds. The ACVM‑0.25% HLC‑I scaffold was revealed to exhibit the best cell proliferation and good cell architecture. ADM and Bio‑Gide scaffolds exhibited good mechanical stability but cell proliferation was reduced when compared with the ACVM‑0.25% HLC‑I scaffold. In addition, SIS scaffolds exhibited the worst cell proliferation. The ACVM‑0.25% HLC‑I scaffold exhibited the best water absorption, followed by the SIS and Bio‑Gide scaffolds, and then the ADM scaffold. In conclusion, the ACVM‑0.25% HLC‑I scaffold has good mechanical properties as a tissue engineering scaffold and the present results suggest that it has better biological characterization when compared with other scaffold types.
Collapse
Affiliation(s)
- Yan-Ling Qiu
- Department of Periodontics and Oral Mucosa, Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xiao Chen
- Department of Orthodontics, Mianyang Stomatological Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Ya-Li Hou
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University and The Key Laboratory of Stomatology, Shijiazhuang, Hebei 050000, P.R. China
| | - Yan-Juan Hou
- Department of Nephrology, Second Hospital, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Song-Bo Tian
- Department of Oral Medicine, Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Yu-He Chen
- Department of Periodontics and Oral Mucosa, Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Yu
- Department of Periodontics and Oral Mucosa, Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Min-Hai Nie
- Department of Periodontics and Oral Mucosa, Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xu-Qian Liu
- Department of Periodontics and Oral Mucosa, Affiliated Stomatology Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
7
|
Li X, Yang S, Wang L, Liu P, Zhao S, Li H, Jiang Y, Guo Y, Wang X. Resveratrol inhibits paclitaxel-induced neuropathic pain by the activation of PI3K/Akt and SIRT1/PGC1α pathway. J Pain Res 2019; 12:879-890. [PMID: 30881098 PMCID: PMC6404678 DOI: 10.2147/jpr.s185873] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) is one of the essential signaling pathways for the development and maintenance of neuropathic pain. Objective To investigate the effect of resveratrol (RES) on paclitaxel-induced neuropathic pain in rats and elucidate the underlying molecular mechanisms. Method Male Sprague Dawley rats were randomly divided into seven groups (n=10/group): Group C, Group P, Group R, Group R+P, Group LY + R+P, Group LY (the specific inhibitor of PI3K), Group E (the specific inhibitor of sirtuin 1 [SIRT1]). Paw withdrawal mechanical threshold (PWT) and thermal withdrawal latency (TWL) were recorded. Mitochondrial histomorphology was performed by transmission electron microscope. PI3K, p-Akt, and t-Akt expressions were tested using immunohistochemistry. Western blot was used to detect p-Akt, t-Akt, SIRT1, and PGC1α expressions. The apoptosis in the striatum, spinal dorsal horns (SDH), and dorsal root ganglions (DRG) tissues was assayed by TUNEL. ELISA was used to detect the contents of IL-β, IL-10, malondialdehyde (MDA), and superoxide dismutase (SOD) in striatum, SDH, and DRG tissues. Results Compared to the control group, PWT and TWL in the P and LY +R+P groups were significantly decreased on 8th and 14th day after paclitaxel administration (P<0.05). The expressions of p-Akt, SIRT1, and PGC1α were decreased in paclitaxel-induced neuropathic rats; however, the expressions of p-Akt, SIRT1, and PGC1α were significantly increased after RES treatment (P<0.05). Furthermore, the expression of p-Akt was decreased by LY294002 (P<0.05), and amount of SIRT1 and PGC1α expression was inhibited by EX-527 (P<0.05). The t-Akt level was not significantly changed in all groups. RES prevented paclitaxel-induced mitochondrial damage by PI3K/Akt. RES improves the pain symptoms of paclitaxel neuralgia rats by increasing the IL-10 and decreasing the expression of IL-1β. RES increases the SOD and reduces the MDA. RES reduces apoptosis by SIRT1/PGC1α signal pathway. Conclusion Our results suggest that RES may inhibit paclitaxel-induced neuropathic pain via PI3K/Akt and SIRT1/PGC1α pathways.
Collapse
Affiliation(s)
- Xiaoning Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Shuhong Yang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Liang Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Peng Liu
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Shuang Zhao
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Huizhou Li
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| | - Yuqing Jiang
- Department of Urology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China
| | - Yuexian Guo
- Department of Urology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China
| | - Xiuli Wang
- Department of Anesthesiology, The Third Hospital of Hebei Medical University, Qiaoxi District, Shijiazhuang 050051, China,
| |
Collapse
|
8
|
Wu M, Wang J, Zhang Y, Liu H, Dong F. Mineralization Induction of Gingival Fibroblasts and Construction of a Sandwich Tissue-Engineered Complex for Repairing Periodontal Defects. Med Sci Monit 2018; 24:1112-1123. [PMID: 29470454 PMCID: PMC5830924 DOI: 10.12659/msm.908791] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The ideal healing technique for periodontal tissue defects would involve the functional regeneration of the alveolar bone, cementum, and periodontal ligament, with new periodontal attachment formation. In this study, gingival fibroblasts were induced and a "sandwich" tissue-engineered complex (a tissue-engineered periodontal membrane between 2 tissue-engineered mineralized membranes) was constructed to repair periodontal defects. We evaluated the effects of gingival fibroblasts used as seed cells on the repair of periodontal defects and periodontal regeneration. MATERIAL AND METHODS Primitively cultured gingival fibroblasts were seeded bilaterally on Bio-Gide collagen membrane (a tissue-engineered periodontal membrane) or unilaterally on small intestinal submucosa segments, and their mineralization was induced. A tissue-engineered sandwich was constructed, comprising the tissue-engineered periodontal membrane flanked by 2 mineralized membranes. Periodontal defects in premolar regions of Beagles were repaired using the tissue-engineered sandwich or periodontal membranes. Periodontal reconstruction was compared to normal and trauma controls 10 or 20 days postoperatively. RESULTS Periodontal defects were completely repaired by the sandwich tissue-engineered complex, with intact new alveolar bone and cementum, and a new periodontal ligament, 10 days postoperatively. CONCLUSIONS The sandwich tissue-engineered complex can achieve ideal periodontal reconstruction rapidly.
Collapse
Affiliation(s)
- Mingxuan Wu
- Department of Oral Medicine, College and Hospital of Stomatology, Hebei Medical University; The Key Laboratory of Stomatology, Shijiazhuang, Hebei, China (mainland)
| | - Jie Wang
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University; The Key Laboratory of Stomatology, Shijiazhuang, Hebei, China (mainland)
| | - Yanning Zhang
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University; The Key Laboratory of Stomatology, Shijiazhuang, Hebei, China (mainland)
| | - Huijuan Liu
- Department of Oral Pathology, College and Hospital of Stomatology, Hebei Medical University; The Key Laboratory of Stomatology, Shijiazhuang, Hebei, China (mainland)
| | - Fusheng Dong
- Department of Oral and Maxillofacial Surgery, College and Hospital of Stomatology, Hebei Medical University; The Key Laboratory of Stomatology, Shijiazhuang, Hebei, China (mainland)
| |
Collapse
|
9
|
Study of composite vascular scaffold combining with differentiated VSMC- and VEC-like cells in vitro and in vivo. J Biomater Appl 2017. [DOI: 10.1177/0885328217715363] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|