1
|
Chen S, Zeng X, Wu M, Zhu J, Wu Y. Sodium Alginate Hydrogel Infusion of Bone Marrow Mesenchymal Stem Cell-Derived Extracellular Vesicles and p38α Antagonistic Peptides in Myocardial Infarction Fibrosis Mitigation. J Am Heart Assoc 2025; 14:e036887. [PMID: 40178108 DOI: 10.1161/jaha.124.036887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/27/2024] [Indexed: 04/05/2025]
Abstract
BACKGROUND Myocardial fibrosis is a pathological hallmark of heart failure post infarction, emphasizing the need for innovative treatment strategies. This research assesses the antifibrotic potential of a sodium alginate (SA) hydrogel loaded with extracellular vesicles (EVs) from bone marrow mesenchymal stem cells and PAP (p38α antagonistic peptides), aiming to interfere with fibrosis-inducing pathways in myocardial tissue after infarction. METHODS We induced fibrosis in mouse cardiac fibroblasts through hypoxia and disrupted the Mapk14 gene to study its contribution to fibrosis. Mesenchymal stem cell-derived EVs, loaded with PAP, were encapsulated in the SA hydrogel (EVs-PAP@SA). The formulation was tested in vitro for its effect on fibrotic marker expression and cell behavior, and in vivo in a murine model of myocardial infarction for its therapeutic efficacy. RESULTS Map k14 silencing showed a decrease in the fibrotic response of cardiac fibroblasts. Treatment with the EVs-PAP@SA hydrogel notably reduced profibrotic signaling, increased cell proliferation and migration, and lowered apoptosis rates. The in vivo treatment with the hydrogel post myocardial infarction significantly diminished myocardial fibrosis and improved cardiac performance. CONCLUSIONS The study endorses the SA hydrogel as an effective vehicle for delivering mesenchymal stem cell-derived EVs and PAP to the heart post myocardial infarction, providing a novel approach for modulating myocardial fibrosis and promoting cardiac healing.
Collapse
Affiliation(s)
- Siyao Chen
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Xiaodong Zeng
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Meifeng Wu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Jiade Zhu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| | - Yijin Wu
- Department of Intensive Care Unit of Cardiovascular Surgery Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University Guangzhou China
| |
Collapse
|
2
|
Silwal P, Nguyen-Thai AM, Alexander PG, Sowa GA, Vo NV, Lee JY. Cellular and Molecular Mechanisms of Hypertrophy of Ligamentum Flavum. Biomolecules 2024; 14:1277. [PMID: 39456209 PMCID: PMC11506588 DOI: 10.3390/biom14101277] [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: 08/29/2024] [Revised: 09/23/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Hypertrophy of the ligamentum flavum (HLF) is a common contributor to lumbar spinal stenosis (LSS). Fibrosis is a core pathological factor of HLF resulting in degenerative LSS and associated low back pain. Although progress has been made in HLF research, the specific molecular mechanisms that promote HLF remain to be defined. The molecular factors involved in the onset of HLF include increases in inflammatory cytokines such as transforming growth factor (TGF)-β, matrix metalloproteinases, and pro-fibrotic growth factors. In this review, we discuss the current understanding of the mechanisms involved in HLF with a particular emphasis on aging and mechanical stress. We also discuss in detail how several pathomechanisms such as fibrosis, proliferation and apoptosis, macrophage infiltration, and autophagy, in addition to several molecular pathways involving TGF-β1, mitogen-activated protein kinase (MAPKs), and nuclear factor-κB (NF-κB) signaling, PI3K/AKT signaling, Wnt signaling, micro-RNAs, extracellular matrix proteins, reactive oxygen species (ROS), etc. are involved in fibrosis leading to HLF. We also present a summary of the current advancements in preclinical animal models for HLF research. In addition, we update the current and potential therapeutic targets/agents against HLF. An improved understanding of the molecular processes behind HLF and a novel animal model are key to developing effective LSS prevention and treatment strategies.
Collapse
Affiliation(s)
- Prashanta Silwal
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Allison M. Nguyen-Thai
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, CA 90095, USA
| | - Peter G. Alexander
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Gwendolyn A. Sowa
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh Medical Cancer, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Nam V. Vo
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Joon Y. Lee
- Ferguson Laboratory for Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
3
|
Liu F, Zhong M, Yang L, Song C, Chen C, Xu Z, Zhang C, Li Z, Wu X, Jiang C, Chen F, Yan Q. Experimental confirmation and bioinformatics reveal biomarkers of immune system infiltration and hypertrophy ligamentum flavum. JOR Spine 2024; 7:e1354. [PMID: 39071860 PMCID: PMC11272949 DOI: 10.1002/jsp2.1354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/23/2024] [Accepted: 06/29/2024] [Indexed: 07/30/2024] Open
Abstract
Background Hypertrophy ligamentum flavum is a prevalent chronic spinal condition that affects middle-aged and older adults. However, the molecular pathways behind this disease are not well comprehended. Objective The objective of this work is to implement bioinformatics techniques in order to identify crucial biological markers and immune infiltration that are linked to hypertrophy ligamentum flavum. Further, the study aims to experimentally confirm the molecular mechanisms that underlie the hypertrophy ligamentum flavum. Methods The corresponding gene expression profiles (GSE113212) were selected from a comprehensive gene expression database. The gene dataset for hypertrophy ligamentum flavum was acquired from GeneCards. A network of interactions between proteins was created, and an analysis of functional enrichment was conducted using the Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) databases. An study of hub genes was performed to evaluate the infiltration of immune cells in patient samples compared to tissues from the control group. Finally, samples of the ligamentum flavum were taken with the purpose of validating the expression of important genes in a clinical setting. Results Overall, 27 hub genes that were differently expressed were found through molecular biology. The hub genes were found to be enriched in immune response, chemokine-mediated signaling pathways, inflammation, ossification, and fibrosis processes, as demonstrated by GO and KEGG studies. The main signaling pathways involved include the TNF signaling pathway, cytokine-cytokine receptor interaction, and TGF-β signaling pathway. An examination of immunocell infiltration showed notable disparities in B cells (naïve and memory) and activated T cells (CD4 memory) between patients with hypertrophic ligamentum flavum and the control group of healthy individuals. The in vitro validation revealed markedly elevated levels of ossification and fibrosis-related components in the hypertrophy ligamentum flavum group, as compared to the normal group. Conclusion The TGF-β signaling pathway, TNF signaling pathway, and related hub genes play crucial roles in the progression of ligamentum flavum hypertrophic. Our study may guide future research on fibrosis of the ligamentum flavum.
Collapse
Affiliation(s)
- Fei Liu
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
- Department of OrthopedicsThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Min Zhong
- Department of ElectrocardiographyThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| | - Lei Yang
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Chao Song
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
- Department of OrthopedicsThe Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical UniversityLuzhouChina
| | - Chaoqi Chen
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Zhiwei Xu
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Chi Zhang
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Zhifa Li
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Xiaofei Wu
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Chen Jiang
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Feng Chen
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| | - Qian Yan
- Department of OrthopedicsRuiKang Hospital affiliated to Guangxi University of Chinese MedicineNanningChina
| |
Collapse
|
4
|
Wang C, Wang Z, Zi Y, Dan X, Xu J, Zhao J, Xu W, Wu Z, Liu W, Ma B. Compensatory upregulation of MT2A alleviates neurogenic intermittent claudication through inhibiting activated p38 MAPK-mediated neuronal apoptosis. Hum Cell 2024; 37:675-688. [PMID: 38546949 DOI: 10.1007/s13577-024-01043-4] [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: 08/28/2023] [Accepted: 02/08/2024] [Indexed: 04/15/2024]
Abstract
Neurogenic intermittent claudication (NIC), a classic symptom of lumbar spinal stenosis (LSS), is associated with neuronal apoptosis. To explore the novel therapeutic target of NIC treatment, we constructed the rat model of NIC by cauda equina compression (CEC) method and collected dorsal root ganglion (DRG) tissues, a region responsible for sensory and motor function, for mRNA sequencing. Bioinformatic analysis of mRNA sequencing indicated that upregulated metallothionein 2A (MT2A), an apoptosis-regulating gene belonging to the metallothionein family, might participate in NIC progression. Activated p38 MAPK mediated motor dysfunction following LSS and it was also found in DRG tissues of rats with NIC. Therefore, we supposed that MT2A might affect NIC progression by regulating p38 MAPK pathway. Then the rat model of NIC was used to explore the exact role of MT2A. Rats at day 7 post-CEC exhibited poorer motor function and had two-fold MT2A expression in DRG tissues compared with rats with sham operation. Co-localization analysis showed that MT2A was highly expressed in neurons, but not in microglia or astrocytes. Subsequently, neurons isolated from DRG tissues of rats were exposed to hypoxia condition (3% O2, 92% N2, 5% CO2) to induce cell damage. Gain of MT2A function in neurons was performed by lentivirus-mediated overexpression. MT2A overexpression inhibited apoptosis by inactivating p38 MAPK in hypoxia-exposed neurons. Our findings indicated that high MT2A expression was related to NIC progression, and MT2A overexpression protected against NIC through inhibiting activated p38 MAPK-mediated neuronal apoptosis in DRG tissues.
Collapse
Affiliation(s)
- Chenggang Wang
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhanchao Wang
- Department of Orthopedics, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Ying Zi
- Department of Orthopedics, Air Force Hospital of the Northern Theater of Chinese People's Liberation Army (PLA), Shenyang, Liaoning, China
| | - Xuejian Dan
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jiahui Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Jingwei Zhao
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wei Xu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Zhourui Wu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wei Liu
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Bin Ma
- Division of Spine, Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
5
|
Sobański D, Bogdał P, Staszkiewicz R, Sobańska M, Filipowicz M, Czepko RA, Strojny D, Grabarek BO. Evaluation of differences in expression pattern of three isoforms of the transforming growth factor beta in patients with lumbosacral stenosis. Cell Cycle 2024; 23:555-572. [PMID: 38695374 PMCID: PMC11135850 DOI: 10.1080/15384101.2024.2345484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 03/30/2024] [Indexed: 05/28/2024] Open
Abstract
The study investigates molecular changes in the lumbosacral (L/S) spine's yellow ligamentum flavum during degenerative stenosis, focusing on the role of transforming growth factor beta 1-3 (TGF-β-1-3). Sixty patients with degenerative stenosis and sixty control participants underwent molecular analysis using real-time quantitative reverse transcription reaction technique (RTqPCR), enzyme-linked immunosorbent assay (ELISA), Western blot, and immunohistochemical analysis (IHC). At the mRNA level, study samples showed reduced expression of TGF-β-1 and TGF-β-3, while TGF-β-2 increased by only 4%. Conversely, at the protein level, the study group exhibited significantly higher concentrations of TGF-β-1, TGF-β-2, and TGF-β-3 compared to controls. On the other hand, at the protein level, a statistically significant higher concentration of TGF-β-1 was observed (2139.33 pg/mL ± 2593.72 pg/mL vs. 252.45 pg/mL ± 83.89 pg/mL; p < 0.0001), TGF-β-2 (3104.34 pg/mL ± 1192.74 pg/mL vs. 258.86 pg/mL ± 82.98 pg/mL; p < 0.0001), TGF-β-3 (512.75 pg/mL ± 107.36 pg/mL vs. 55.06 pg/mL ± 9.83 pg/mL, p < 0.0001) in yellow ligaments obtained from patients of the study group compared to control samples. The study did not establish a significant correlation between TGF-β-1-3 concentrations and pain severity. The findings suggest that molecular therapy aimed at restoring the normal expression pattern of TGF-β-1-3 could be a promising strategy for treating degenerative stenosis of the L/S spine. The study underscores the potential therapeutic significance of addressing molecular changes at the TGF-β isoforms level for better understanding and managing degenerative spinal conditions.
Collapse
Affiliation(s)
- Dawid Sobański
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
| | - Paweł Bogdał
- Department of Orthopedic, Szpital Powiatowy w Zawierciu, Zawiercie, Poland
| | - Rafał Staszkiewicz
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
- Department of Neurosurgery, 5th Military Clinical Hospital with the SP ZOZ Polyclinic in Krakow, Krakow, Poland
- Department of Neurosurgery, Faculty of Medicine in Zabrze, Academy of Silesia, Katowice, Poland
| | | | - Michał Filipowicz
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
| | - Ryszard Adam Czepko
- Department of Neurosurgery, Szpital sw. Rafala in Cracow, Cracow, Poland
- Department of Neurosurgery, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University in Cracow, Cracow, Poland
| | - Damian Strojny
- Collegium Medicum, WSB University, Dabrowa Gornicza, Poland
- Institute of Health Care, National Academy of Applied Sciences in Przemyśl, Przemyśl, Poland
- Department of Medical Science, New Medical Techniques Specialist Hospital of St. Family in Rudna Mała, Rzeszów, Poland
| | | |
Collapse
|
6
|
Goto Y, Kato K, Yagi K, Kawaguchi Y, Yonezu H, Koshimae T, Waguri-Nagaya Y, Murakami H, Suzuki N. Transforming Growth Factor-β Induces Interleukin-6 Secretion from Human Ligamentum Flavum-Derived Cells through Partial Activation of p38 and p44/42 Mitogen-Activated Protein Kinases. Asian Spine J 2023; 17:997-1003. [PMID: 37946333 PMCID: PMC10764132 DOI: 10.31616/asj.2023.0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/03/2023] [Accepted: 04/23/2023] [Indexed: 11/12/2023] Open
Abstract
STUDY DESIGN This experimental study was performed using human ligamentum flavum-derived cells (HFCs). PURPOSE To investigate the intracellular signaling mechanism of interleukin-6 (IL-6) secretion in transforming growth factor-β (TGF- β)-stimulated HFCs. OVERVIEW OF LITERATURE Lumbar spinal stenosis (LSS) is a prevalent disease among the elderly, characterized by debilitating pain in the lower extremities. Although the number of patients with LSS has increased in recent years, the underlying pathomechanism remains unclear. Clinical examinations typically rely on magnetic resonance imaging to diagnose patients, revealing ligamentum flavum hypertrophy. Some studies have suggested an association between ligamentum flavum hypertrophy and inflammation/fibrosis, and expression of TGF-β and IL-6 has been observed in surgically obtained ligamentum flavum samples. However, direct evidence linking TGF-β and IL-6 expression in HFCs is lacking. METHODS HFCs were obtained from patients with LSS who had undergone decompression surgery. The cells were stimulated with TGF-β and pretreated with either the p38 mitogen-activated protein (MAP) kinase inhibitor SB203580 or the p44/42 MAP kinase inhibitor FR180204. IL-6 secretion in the cell culture medium and IL-6 messenger RNA (mRNA) expression levels were analyzed using an enzyme-linked immunoassay and real-time polymerase chain reaction, respectively. RESULTS TGF-β administration resulted in a dose- and time-dependent stimulation of IL-6 release. Treatment with SB203580 and FR180204 markedly suppressed TGF-β-induced IL-6 secretion from HFCs. Moreover, these inhibitors suppressed IL-6 mRNA expression in response to TGF-β stimulation. CONCLUSIONS Our findings indicate that TGF-β induces IL-6 protein secretion and gene expression in HFCs through the activation of p38 or p44/42 MAP kinases. These results suggest a potential association between IL-6-mediated inflammatory response and tissue hypertrophy in LSS, and we provide insights into molecular targets for therapeutic interventions targeting LSS-related inflammation through our analysis of the MAP kinase pathway using HFCs.
Collapse
Affiliation(s)
- Yuta Goto
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Kenji Kato
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Kiyoshi Yagi
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Yohei Kawaguchi
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Hiroki Yonezu
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Tomoko Koshimae
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Yuko Waguri-Nagaya
- Department of Orthopaedic Surgery, Nagoya City University East Medical Center, Nagoya,
Japan
| | - Hideki Murakami
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| | - Nobuyuki Suzuki
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya,
Japan
| |
Collapse
|
7
|
Davies BM, Banerjee A, Mowforth OD, Kotter MRN, Newcombe VFJ. Is the type and/or co-existence of degenerative spinal pathology associated with the occurrence of degenerative cervical myelopathy? A single centre retrospective analysis of individuals with MRI defined cervical cord compression. J Clin Neurosci 2023; 117:84-90. [PMID: 37783068 DOI: 10.1016/j.jocn.2023.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/13/2023] [Accepted: 09/17/2023] [Indexed: 10/04/2023]
Abstract
BACKGROUND Degenerative cervical myelopathy (DCM) arises from spinal degenerative changes injuring the cervical spinal cord. Most cord compression is incidental, referred to as asymptomatic spinal cord compression (ASCC). How and why ASCC differs from DCM is poorly understood. In this paper, we study a local cohort to identify specific types and groups of degenerative pathology more likely associated with DCM than ASCC. METHODS This study was a retrospective cohort analysis (IRB Approval ID: PRN10455). The frequency of degenerative findings between those with ASCC and DCM patients were compared using network analysis, hierarchical clustering, and comparison to existing literature to identify potential subgroups in a local cohort (N = 155) with MRI-defined cervical spinal cord compression. Quantitative measures of spinal cord compression (MSCC and MCC) were used to confirm their relevance. RESULTS ELF (8.7 %, 95 % CI 3.8-13.6 % vs 35.7 %, 95 % CI 27.4-44.0 %) Congenital Stenosis (3.9 %, 95 % CI 0.6-7.3 % vs 25.0 %, 95 % CI 17.5-32.5 %), and OPLL (0.0 %, 95 % CI 0.0-0.0 % vs 3.6 %, 95 % CI 0.3-6.8 %) were more likely in patients with DCM. Comparative network analysis indicated loss of lordosis was associated with ASCC, whilst ELF with DCM. Hierarchical Cluster Analysis indicated four sub-groups: multi-level disc disease with ELF, single-level disc disease without loss of lordosis and OPLL with DCM, and single-level disc disease with loss of lordosis with ASCC. Quantitative measures of cord compression were higher in groups associated with DCM, but similar in patients with single-level disc disease and loss of lordosis. CONCLUSIONS This study identified four subgroups based on degenerative pathology requiring further investigation.
Collapse
Affiliation(s)
- Benjamin M Davies
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, UK.
| | - Arka Banerjee
- St George's University Hospitals NHS Foundation Trust, London, UK
| | - Oliver D Mowforth
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, UK
| | - Mark R N Kotter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, UK
| | | |
Collapse
|
8
|
Shin HK, Seo KJ, Lee JY, Jeon SR, Yune TY. GSK-3β and β-Catenin Signaling Pathway is Involved in Myofibroblast Transition of Ligamentum Flavum in Lumbar Spinal Stenosis Patients. Spine (Phila Pa 1976) 2023; 48:1472-1479. [PMID: 37417723 DOI: 10.1097/brs.0000000000004770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 06/25/2023] [Indexed: 07/08/2023]
Abstract
STUDY DESIGN Histologic analysis of the ligamentum flavum (LF) in the lumbar spine. OBJECTIVE The objective of this study is to investigate the levels of glycogen synthase kinase-3β (GSK-3β) and β-catenin in the LF tissue of patients with lumbar spinal stenosis (LSS). SUMMARY OF BACKGROUND DATA The hypertrophy of the LF is the primary cause of the progression of LSS. Recently, Wnt signaling has been proposed as one of the molecular processes contributing to LF hypertrophy. GSK-3β and β-catenin are recognized to play a crucial part in the control of this signaling pathway. MATERIALS AND METHODS From May 2020 to July 2022, LF from 51 LSS patients (LSS group) and 18 lumbar disc herniation patients (control group) were prospectively collected during surgery. Histologic analysis was investigated to confirm the progression of LF fibrosis. The levels of α-smooth muscle actin, phosphorylation of GSK-3β (p-GSK-3β; inactive form), and β-catenin were analyzed in LF with Western blot analysis to reveal the GSK-3β/β-catenin signaling pathway. Continuous variables are expressed as mean±SD and compared using the student t test. Categorical variables are compared using the χ 2 test or Fisher exact test, as appropriate. To determine the association between p-GSK-3β and LF thickness, the Pearson correlation coefficient was calculated based on the results of Western blot analysis. RESULTS The LSS group was older and had thicker LF than the controls. The LSS group showed increased collagen fiber and cellularity than the controls. The levels of α-smooth muscle actin, p-GSK-3β, and β-catenin in the LF of the LSS group were significantly higher than that of the control group. There was a strong positive correlation between p-GSK-3β (Ser9) level and LF thickness in LSS patients ( r =0.69, P =0.01). CONCLUSION This research proposes a molecular mechanism for the pathogenesis of LF hypertrophy in LSS. Specifically, GSK-3β/β-catenin signaling appears to be related to LF hypertrophy in LSS and a positive correlation exists between p-GSK-3β level and LF thickness. LEVEL OF EVIDENCE Level 3.
Collapse
Affiliation(s)
- Hong Kyung Shin
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Jin Seo
- Department Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul, Republic of Korea
| | - Jee Youn Lee
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Ryong Jeon
- Department of Neurological Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Young Yune
- Department Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul, Republic of Korea
- Biomedical Science Institute, Kyung Hee University, Seoul, Republic of Korea
- Age-Related and Brain Diseases Research Center, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Hassan MDS, Razali N, Abu Bakar AS, Abu Hanipah NF, Agarwal R. Connective tissue growth factor: Role in trabecular meshwork remodeling and intraocular pressure lowering. Exp Biol Med (Maywood) 2023; 248:1425-1436. [PMID: 37873757 PMCID: PMC10657592 DOI: 10.1177/15353702231199466] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023] Open
Abstract
Connective tissue growth factor (CTGF) is a distinct signaling molecule modulating many physiological and pathophysiological processes. This protein is upregulated in numerous fibrotic diseases that involve extracellular matrix (ECM) remodeling. It mediates the downstream effects of transforming growth factor beta (TGF-β) and is regulated via TGF-β SMAD-dependent and SMAD-independent signaling routes. Targeting CTGF instead of its upstream regulator TGF-β avoids the consequences of interfering with the pleotropic effects of TGF-β. Both CTGF and its upstream mediator, TGF-β, have been linked with the pathophysiology of glaucomatous optic neuropathy due to their involvement in the regulation of ECM homeostasis. The excessive expression of these growth factors is associated with glaucoma pathogenesis via elevation of the intraocular pressure (IOP), the most important risk factor for glaucoma. The raised in the IOP is due to dysregulation of ECM turnover resulting in excessive ECM deposition at the site of aqueous humor outflow. It is therefore believed that CTGF could be a potential therapeutic target in glaucoma therapy. This review highlights the CTGF biology and structure, its regulation and signaling, its association with the pathophysiology of glaucoma, and its potential role as a therapeutic target in glaucoma management.
Collapse
Affiliation(s)
| | - Norhafiza Razali
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Amy Suzana Abu Bakar
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Noor Fahitah Abu Hanipah
- Institute of Medical Molecular Biotechnology (IMMB), Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi MARA (UiTM), 47000 Sungai Buloh, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University (IMU), 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
10
|
Hsu YC, Chuang HC, Tsai KL, Tu TY, Shyong YJ, Kuo CH, Liu YF, Shih SS, Lin CL. Administration of N-Acetylcysteine to Regress the Fibrogenic and Proinflammatory Effects of Oxidative Stress in Hypertrophic Ligamentum Flavum Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1380353. [PMID: 36338342 PMCID: PMC9629932 DOI: 10.1155/2022/1380353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/22/2022] [Accepted: 09/20/2022] [Indexed: 03/22/2025]
Abstract
Ligamentum flavum hypertrophy (LFH) is a major cause of lumbar spinal stenosis (LSS). In hypertrophic ligamentum flavum (LF) cells, oxidative stress activates intracellular signaling and induces the expression of inflammatory and fibrotic markers. This study explored whether healthy and hypertrophic LF cells respond differently to oxidative stress, via examining the levels of phosphorylated p38 (p-p38), inducible nitric oxide synthase (iNOS), and α-smooth muscle actin (α-SMA). Furthermore, the efficacy of N-acetylcysteine (NAC), an antioxidant, in reversing the fibrogenic and proinflammatory effects of oxidative stress in hypertrophic LF cells was investigated by assessing the expression levels of p-p38, p-p65, iNOS, TGF-β, α-SMA, vimentin, and collagen I under H2O2 treatment with or without NAC. Under oxidative stress, p-p38 increased significantly in both hypertrophic and healthy LF cells, and iNOS was elevated in only the hypertrophic LF cells. This revealed that oxidative stress negatively affected both hypertrophic and healthy LF cells, with the hypertrophic LF cells exhibiting more active inflammation than did the healthy cells. After H2O2 treatment, p-p38, p-p65, iNOS, TGF-β, vimentin, and collagen I increased significantly, and NAC administration reversed the effects of oxidative stress. These results can form the basis of a novel therapeutic treatment for LFH using antioxidants.
Collapse
Affiliation(s)
- Yu-Chia Hsu
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hao-Chun Chuang
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kun-Ling Tsai
- Department of Physical Therapy, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Yan-Jye Shyong
- Department of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Cheng-Hsiang Kuo
- Department of Biochemistry and Molecular Biology, National Cheng Kung University, Tainan, Taiwan
| | - Yuan-Fu Liu
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Shien Shih
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Li Lin
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
11
|
The Discovery and Development of Natural-Based Biomaterials with Demonstrated Wound Healing Properties: A Reliable Approach in Clinical Trials. Biomedicines 2022; 10:biomedicines10092226. [PMID: 36140332 PMCID: PMC9496351 DOI: 10.3390/biomedicines10092226] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 09/04/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Current research across the globe still focuses strongly on naturally derived biomaterials in various fields, particularly wound care. There is a need for more effective therapies that will address the physiological deficiencies underlying chronic wound treatment. The use of moist bioactive scaffolds has significantly increased healing rates compared to local and traditional treatments. However, failure to heal or prolonging the wound healing process results in increased financial and social stress imposed on health institutions, caregivers, patients, and their families. The urgent need to identify practical, safe, and cost-effective wound healing scaffolding from natural-based biomaterials that can be introduced into clinical practice is unequivocal. Naturally derived products have long been used in wound healing; however, clinical trial evaluations of these therapies are still in their infancy. Additionally, further well-designed clinical trials are necessary to confirm the efficacy and safety of natural-based biomaterials in treating wounds. Thus, the focus of this review is to describe the current insight, the latest discoveries in selected natural-based wound healing implant products, the possible action mechanisms, and an approach to clinical studies. We explore several tested products undergoing clinical trials as a novel approach to counteract the debilitating effects of impaired wound healing.
Collapse
|
12
|
Banerjee A, Mowforth OD, Nouri A, Budu A, Newcombe V, Kotter MRN, Davies BM. The Prevalence of Degenerative Cervical Myelopathy-Related Pathologies on Magnetic Resonance Imaging in Healthy/Asymptomatic Individuals: A Meta-Analysis of Published Studies and Comparison to a Symptomatic Cohort. J Clin Neurosci 2022; 99:53-61. [PMID: 35255357 DOI: 10.1016/j.jocn.2022.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 11/29/2022]
Abstract
Degenerative cervical myelopathy (DCM) is a progressive cervical spinal cord injury brought about by mechanical stress from degenerative changes in the cervical spine. It is typically diagnosed on clinical symptoms and examination findings together with MRI findings. In this study, we explore the significance of these degenerative pathology to onset of DCM by performing the first meta-analysis on the prevalence of degenerative features reported on MRI amongst healthy and asymptomatic populations and compare this to the prevalence of degenerative features reported on MRI amongst a symptomatic population calculated in a previous review. We conducted a systematic review and meta-analysis in accordance with PRISMA guidelines, cognizant of their adaptation for epidemiological studies. A search strategy was used to identify original research carrying out MRI screening of cervical spines of asymptomatic patients in MEDLINE and Embase from 1985 to present day. The search yielded a total of 1098 studies of which 17 were included in this meta-analysis covering a total of 5059 patients. Ossification of posterior longitudinal ligament (pooled asymptomatic prevalence of 0.4%, 95% Confidence Interval [0.1%, 0.8%]), enlargement of ligamentum flavum (pooled asymptomatic prevalence of 11.8%, 95% Confidence Interval [5.3%, 18.4%]) and degenerative multilevel disc pathology (pooled asymptomatic prevalence of 64.5%, 95% Confidence Interval [48.3%, 80.8%], I2 100%) were found to be significantly lower in asymptomatic populations. Symptomatic populations have a prevalence of 10.5% (95% Confidence Interval [7.7%, 13.3%]) for ossification of posterior longitudinal ligament, 56.8% (95% Confidence Interval [52.3%, 61.3%]) for enlargement of ligamentum flavum and 89.7% (95% Confidence Interval [86.9%, 92.5%]) for degenerative multilevel disc pathology [18]. Understanding the natural history of DCM is a recognised research priority, and whilst these perspectives require further evaluation, they may be of significant relevance to the evolving biomechanical understanding of the disease.
Collapse
Affiliation(s)
| | | | - Aria Nouri
- Department of Neurosurgery, University of Geneva, 1205 Geneva, Switzerland
| | - Alexandru Budu
- Department of Neurosurgery, Queen Elizabeth Hospital, Birmingham, UK
| | - Virginia Newcombe
- Division of Anaesthesia, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Mark R N Kotter
- Department of Neurosurgery, University of Cambridge, Cambridge, UK; Myelopathy.org, University of Cambridge, UK
| | | |
Collapse
|
13
|
Yang K, Chen Y, Xiang X, Lin Y, Fei C, Chen Z, Lai Z, Yu Y, Tan R, Dong J, Zhang J, Li P, Wang L, Zhang Z. EGF Contributes to Hypertrophy of Human Ligamentum Flavum via the TGF-β1/Smad3 Signaling Pathway. Int J Med Sci 2022; 19:1510-1518. [PMID: 36185336 PMCID: PMC9515692 DOI: 10.7150/ijms.76077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Background: The most common spinal disorder in elderly is lumbar spinal canal stenosis (LSCS). Previous studies showed that ligamentum flavum hypertrophy (LFH) with fibrosis as the main pathological change is one of the pathogenic factors leading to LSCS. Epidermal Growth Factor (EGF) is known to have an intimate relationship with fibrosis in various tissues. Nevertheless, currently, there are few studies regarding EGF in LFH. The effect of EGF on the development of LFH is unknown, and the underlying pathomechanism remains unclear. In this study, we investigated the role of EGF in LFH and its potential molecular mechanism. Methods: First, the expression levels of EGF, phosphorylation of EGF receptor (pEGFR), Transforming growth factor-β1 (TGF-β1), Phosphorylated Smad3 (pSmad3), collagen I and collagen III were examined via immunohistochemistry and Western blot in LF tissues from patients with LSCS or Non-LSCS. Second, primary LF cells were isolated from adults with normal LF thickness and were cultured with different concentrations of exogenous EGF with or without erlotinib/TGF-β1-neutralizing antibody. Results: The results showed that EGF, pEGFR, TGF-β1, pSmad3, collagen I and collagen III protein expression in the LSCS group was significantly higher than that in the Non-LSCS group. Meanwhile, pEGFR, TGF-β1, pSmad3, collagen I and collagen III protein expression was significantly enhanced in LF cells after exogenous EGF exposure, which can be notably blocked by erlotinib. In addition, pSmad3, collagen I and collagen III protein expression was blocked by TGF-β1-neutralizing antibody. Conclusions: EGF promotes the synthesis of collagen I and collagen III via the TGF-β1/Smad3 signaling pathway, which eventually contributes to LFH.
Collapse
Affiliation(s)
- Kaifan Yang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanlin Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xin Xiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China.,The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yanling Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chengshuo Fei
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zesen Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongming Lai
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongpeng Yu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruiqian Tan
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiale Dong
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junxiong Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Li
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Academy of Orthopedics, Guangzhou, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
14
|
Mechanical Stress-Induced IGF-1 Facilitates col-I and col-III Synthesis via the IGF-1R/AKT/mTORC1 Signaling Pathway. Stem Cells Int 2021; 2021:5553676. [PMID: 34912459 PMCID: PMC8668368 DOI: 10.1155/2021/5553676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/13/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
Mechanical stress promotes human ligamentum flavum cells (LFCs) to synthesize multitype collagens, leading to ligamentum flavum hypertrophy (LFH). However, the mechanism of mechanical stress in the formation of collagen remains unclear. Therefore, we investigated the relationship between mechanical stress and collagen synthesis in the present study. First, LFCs were isolated from 9 patients and cultured with or without mechanical stress exposure for different times. IGF-1, collagen I (col-I), and collagen III (col-III) protein and mRNA levels were then detected via ELISA and qPCR, respectively. Moreover, the activation of pIGF-1R, pAKT, and pS6 was examined by Western blot analysis. To further explore the underlying mechanism, an IGF-1 neutralizing antibody, NVP-AEW541, and rapamycin were used. IGF-1, col-I, and col-III were significantly increased in stressed LFCs compared to nonstressed LFCs. In addition, the activation of pIGF-1R, pAKT, and pS6 was obviously enhanced in stressed LFCs. Interestingly, col-I protein, col-I mRNA, col-III protein, col-III mRNA, and IGF-1 protein, but not IGF-1 mRNA, were inhibited by IGF-1 neutralizing antibody. In addition, col-I and col-III protein and mRNA, but not IGF-1, were inhibited by both NVP-AEW541 and rapamycin. Moreover, the activation of pIGF-1R, pAKT, and pS6 was reduced by the IGF-1 neutralizing antibody and NVP-AEW541, and the activation of pS6 was reduced by rapamycin. In summary, these results suggested that mechanical stress promotes LFCs to produce IGF-1, which facilitates col-I and col-III synthesis via the IGF-1R/AKT/mTORC1 signaling pathway.
Collapse
|
15
|
Cao Y, Zhan Y, Qiu S, Chen Z, Gong K, Ni S, Duan Y. Integrative analysis of genome-wide DNA methylation and single-nucleotide polymorphism identified ACSM5 as a suppressor of lumbar ligamentum flavum hypertrophy. Arthritis Res Ther 2021; 23:251. [PMID: 34593020 PMCID: PMC8482693 DOI: 10.1186/s13075-021-02625-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 09/12/2021] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Hypertrophy of ligamentum flavum (HLF) is a common lumbar degeneration disease (LDD) with typical symptoms of low back pain and limb numbness owing to an abnormal pressure on spinal nerves. Previous studies revealed HLF might be caused by fibrosis, inflammatory, and other bio-pathways. However, a global analysis of HLF is needed severely. METHODS A genome-wide DNA methylation and single-nucleotide polymorphism analysis were performed from five LDD patients with HLF and five LDD patients without HLF. Comprehensive integrated analysis was performed using bioinformatics analysis and the validated experiments including Sanger sequencing, methylation-specific PCR, qPCR and ROC analysis. Furthermore, the function of novel genes in ligamentum flavum cells (LFCs) was detected to explore the molecular mechanism in HLF through knock down experiment, overexpression experiment, CCK8 assay, apoptosis assay, and so on. RESULTS We identified 69 SNP genes and 735 661 differentially methylated sites that were enriched in extracellular matrix, inflammatory, and cell proliferation. A comprehensive analysis demonstrated key genes in regulating the development of HLF including ACSM5. Furthermore, the hypermethylation of ACSM5 that was mediated by DNMT1 led to downregulation of ACSM5 expression, promoted the proliferation and fibrosis, and inhibited the apoptosis of LFCs. CONCLUSION This study revealed that DNMT1/ACSM5 signaling could enhance HLF properties in vitro as a potential therapeutic strategy for HLF.
Collapse
Affiliation(s)
- Yanlin Cao
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yenan Zhan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Sujun Qiu
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Zhong Chen
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Kaiqin Gong
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Songjia Ni
- Department of Orthopaedic Trauma, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| | - Yang Duan
- Department of Spine Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
16
|
Long-term, Time-course Evaluation of Ligamentum Flavum Hypertrophy Induced by Mechanical Stress: An Experimental Animal Study. Spine (Phila Pa 1976) 2021; 46:E520-E527. [PMID: 33273443 DOI: 10.1097/brs.0000000000003832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Experimental animal study. OBJECTIVE The aim of this study was to clarify chronological effects of mechanical stress on ligamentum flavum (LF) using a long-term fusion rabbit model. SUMMARY OF BACKGROUND DATA LF hypertrophy is a major pathology of lumbar spinal stenosis (LSS), but its mechanism remains unclear. We previously demonstrated mechanical-stress-induced LF hypertrophy with a rabbit model. However, we only investigated LFs at a single time point in the short-term; the effects of long-term mechanical stress have not been elucidated. METHODS Eighteen-week-old male New Zealand White rabbits were randomly divided into two groups: the mechanical stress group underwent L2-3 and L4-5 posterolateral fusion and resection of the L3-4 supraspinal muscle, whereas the control group underwent only surgical exposure. Rabbits were sacrificed 16 and 52 weeks after the procedure. Axial specimens of LFs at L3-4 were evaluated histologically. Immunohistochemistry for alpha-smooth muscle actin (α-SMA) was performed to assess the numbers of vessels and myofibroblasts. RESULTS In the mechanical stress group, LFs at the L3-4 level exhibited hypertrophy with elastic fiber disruption and cartilage matrix production at 16 and 52 weeks. A trend test indicated that mechanical stress induced LF hypertrophy, elastic fiber disruption, and cartilage matrix production in a time-dependent manner, with the lowest levels before treatment and the highest at 52 weeks. Immunostaining for α-SMA showed similar numbers of vessels in both groups, whereas the percentage of myofibroblasts was significantly larger at 16 and 52 weeks in the mechanical stress group than in the control group. CONCLUSION We demonstrated that long-term mechanical stress caused LF hypertrophy with progressive elastic fiber disruption and cartilage matrix production accompanied by enhanced myofibroblasts. In addition, the reported rabbit model could be extended to elucidate the mechanism of LF hypertrophy and to develop new therapeutic strategies for LSS by preventing LF hypertrophy.Level of Evidence: SSSSS.
Collapse
|
17
|
Abstract
Hypertrophy of the ligamentum flavum (LF) is a major cause of lumbar spinal stenosis (LSS), and the pathology involves disruption of elastic fibers, fibrosis with increased cellularity and collagens, and/or calcification. Previous studies have implicated the increased expression of the proteoglycan family in hypertrophied LF. Furthermore, the gene expression profile in a rabbit experimental model of LF hypertrophy revealed that biglycan (BGN) is upregulated in hypertrophied LF by mechanical stress. However, the expression and function of BGN in human LF has not been well elucidated. To investigate the involvement of BGN in the pathomechanism of human ligamentum hypertrophy, first we confirmed increased expression of BGN by immunohistochemistry in the extracellular matrix of hypertrophied LF of LSS patients compared to LF without hypertrophy. Experiments using primary cell cultures revealed that BGN promoted cell proliferation. Furthermore, BGN induces changes in cell morphology and promotes myofibroblastic differentiation and cell migration. These effects are observed for both cells from hypertrophied and non-hypertrophied LF. The present study revealed hyper-expression of BGN in hypertrophied LF and function of increased proteoglycan in LF cells. BGN may play a crucial role in the pathophysiology of LF hypertrophy through cell proliferation, myofibroblastic differentiation, and cell migration.
Collapse
|
18
|
Han Y, Wang J, Jin M, Jia L, Yan C, Wang Y. Shentong Zhuyu Decoction Inhibits Inflammatory Response, Migration, and Invasion and Promotes Apoptosis of Rheumatoid Arthritis Fibroblast-like Synoviocytes via the MAPK p38/PPAR γ/CTGF Pathway. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6187695. [PMID: 33511203 PMCID: PMC7826240 DOI: 10.1155/2021/6187695] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The current study is aimed at exploring the effect of Shentong Zhuyu Decoction on the proliferation, migration, invasion, and apoptosis of rheumatoid arthritis fibroblast-like synoviocytes (RA-FLS) and its underlying molecular mechanism. MATERIALS AND METHODS The type II collagen-induced arthritis (CIA) model was established. Subsequently, the RA-FLS were isolated from the CIA rat model and identified by immunohistochemistry. The viability, apoptosis, cell cycle, migration, and invasion of RA-FLS were detected by the cell counting kit 8 (CCK-8) assay, flow cytometry, wound-healing assay, and transwell invasion assay, respectively. The levels of MAPK p38, PPARγ, CTGF, Bcl-2, Bax, caspase-3, IL-1β, MMP-3, CDK4, and cyclin D1 were determined by qRT-PCR and western blotting, respectively. RESULTS After treatment with Shentong Zhuyu Decoction medicated serum, the OD570 value, migrative and invasive abilities, and the secretion of IL-1β, MMP-3 were remarkably decreased in RA-FLS, while the apoptosis rate was increased. Further, results showed that Shentong Zhuyu Decoction inhibited the transition from the G1 phase to S phase. Additionally, Shentong Zhuyu Decoction significantly inhibited the expression of Bcl-2, CDK4, cyclin D1, MAPK p-p38, and CTGF, whereas elevated the levels of Bax, caspase-3, and PPARγ. Importantly, the effects of Shentong Zhuyu Decoction were consistent with the trends of MAPK P38 inhibitor (SB203580) and PPARγ agonist (GW1929). CONCLUSIONS Shentong Zhuyu Decoction inhibited viability, inflammatory response, migration, invasion, and transition from the G1 phase to S phase and promoted apoptosis of RA-FLS via the MAPK p38/PPARγ/CTGF pathway.
Collapse
Affiliation(s)
- Ying Han
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
- Department of Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jing Wang
- Department of Chinese Medicine Diagnostics, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Meng Jin
- Department of Chinese Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lin Jia
- Department II of Respiratory, Hebei Provincial Hospital of Traditional Chinese Medicine, Shijiazhuang, China
| | - Cuihuan Yan
- Institute of Integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Yali Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
19
|
Yagi K, Goto Y, Kato K, Suzuki N, Kondo A, Waseda Y, Mizutani J, Kawaguchi Y, Joyo Y, Waguri-Nagaya Y, Murakami H. p38 Mitogen-Activated Protein Kinase Is Involved in Interleukin-6 Secretion from Human Ligamentum Flavum-Derived Cells Stimulated by Tumor Necrosis Factor-α. Asian Spine J 2020; 15:713-720. [PMID: 33355843 PMCID: PMC8696066 DOI: 10.31616/asj.2020.0425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 10/04/2020] [Indexed: 12/26/2022] Open
Abstract
Study Design Human ligamentum flavum–derived cells (HFCs) were obtained from surgical samples for a basic experimental study. Purpose We sought to evaluate the inflammatory response of human ligamentum flavum cells to investigate hypertrophic changes occurring in the ligamentum flavum. Overview of Literature Lumbar spinal stenosis (LSS) is a disease commonly observed in the elderly. The number of patients with LSS has increased over time, yet the pathomechanisms of LSS still have not been fully elucidated. One of the clinical features of LSS is hypertrophy of the ligamentum flavum, which results in narrowing of the lumbar spinal canal. Some reports have suggested that ligamentum flavum hypertrophy is associated with inflammation and fibrosis; meanwhile, the p38 mitogen-activated protein (MAP) kinase is involved in the hypertrophy of human ligamentum flavum cells. Methods HFCs were obtained from patients with LSS who underwent surgery. HFCs were stimulated by tumor necrosis factor-α (TNF-α) and a p38 MAP kinase inhibitor, SB203580. Phosphorylation of the p38 MAP kinase was analyzed by western blotting. The concentration of interleukin-6 (IL-6) in the conditioned medium was measured by enzyme-linked immunoassay and IL-6 messenger RNA expression levels were determined by real-time polymerase chain reaction. Results TNF-α induced the phosphorylation of p38 MAP kinase in a time-dependent manner, which was suppressed by the p38 MAP kinase inhibitor, SB203580. TNF-α also stimulated IL-6 release in both a time- and dose-dependent manner. On its own, SB203580 did not stimulate IL-6 secretion from HFCs; however, it dramatically suppressed the degree of IL-6 release stimulated by TNF-α from HFCs. Conclusions This is the first report suggesting that TNF-α stimulates the gene expression and protein secretion of IL-6 via p38 MAP kinase in HFCs. A noted association between tissue hypertrophy and inflammation suggests that the p38 MAP kinase inflammatory pathway may be a therapeutic molecular target for LSS.
Collapse
Affiliation(s)
- Kiyoshi Yagi
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuta Goto
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kenji Kato
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Nobuyuki Suzuki
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akira Kondo
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuya Waseda
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Jun Mizutani
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yohei Kawaguchi
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuji Joyo
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuko Waguri-Nagaya
- Department of Orthopaedic Surgery, Nagoya City East Medical Center, Nagoya, Japan
| | - Hideki Murakami
- Department of Orthopaedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
20
|
Lin CL, Kuo YT, Tsao CH, Shyong YJ, Shih SH, Tu TY. Development of an In Vitro 3D Model for Investigating Ligamentum Flavum Hypertrophy. Biol Proced Online 2020; 22:20. [PMID: 32884451 PMCID: PMC7460798 DOI: 10.1186/s12575-020-00132-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/29/2020] [Indexed: 12/22/2022] Open
Abstract
Background Ligamentum flavum hypertrophy (LFH) is among the most crucial factors in degenerative lumbar spinal stenosis, which can cause back pain, lower extremity pain, cauda equina syndrome and neurogenic claudication. The exact pathogenesis of LFH remains elusive despite extensive research. Most in vitro studies investigating LFH have been carried out using conventional two-dimensional (2D) cell cultures, which do not resemble in vivo conditions, as they lack crucial pathophysiological factors found in three-dimensional (3D) LFH tissue, such as enhanced cell proliferation and cell cluster formation. In this study, we generated ligamentum flavum (LF) clusters using spheroid cultures derived from primary LFH tissue. Results The cultured LF spheroids exhibited good viability and growth on an ultra-low attachment 96-well plate (ULA 96-plate) platform according to live/dead staining. Our results showed that the 100-cell culture continued to grow in size, while the 1000-cell culture maintained its size, and the 5000-cell culture exhibited a decreasing trend in size as the culture time increased; long-term culture was validated for at least 28 days. The LF spheroids also maintained the extracellular matrix (ECM) phenotype, i.e., fibronectin, elastin, and collagen I and III. The 2D culture and 3D culture were further compared by cell cycle and Western blot analyses. Finally, we utilized hematoxylin and eosin (H&E) staining to demonstrate that the 3D spheroids resembled part of the cell arrangement in LF hypertrophic tissue. Conclusions The developed LF spheroid model has great potential, as it provides a stable culture platform in a 3D model that can further improve our understanding of the pathogenesis of LFH and has applications in future studies.
Collapse
Affiliation(s)
- Cheng-Li Lin
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Skeleton Materials and Bio-compatibility Core Lab, Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan.,Medical Device Innovation Center (MDIC), National Cheng Kung University, Tainan, 70101 Taiwan
| | - Yi-Ting Kuo
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Che-Hao Tsao
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Yan-Jye Shyong
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101 Taiwan.,Institute of Clinical Pharmacy and Pharmaceutical Sciences, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Shu-Hsien Shih
- Department of Orthopaedic Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Ting-Yuan Tu
- Medical Device Innovation Center (MDIC), National Cheng Kung University, Tainan, 70101 Taiwan.,Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101 Taiwan.,International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, 70101 Taiwan
| |
Collapse
|
21
|
Sun C, Zhang H, Wang X, Liu X. Ligamentum flavum fibrosis and hypertrophy: Molecular pathways, cellular mechanisms, and future directions. FASEB J 2020; 34:9854-9868. [PMID: 32608536 DOI: 10.1096/fj.202000635r] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/11/2022]
Abstract
Hypertrophy of ligamentum flavum (LF), along with disk protrusion and facet joints degeneration, is associated with the development of lumbar spinal canal stenosis (LSCS). Of note, LF hypertrophy is deemed as an important cause of LSCS. Histologically, fibrosis is proved to be the main pathology of LF hypertrophy. Despite the numerous studies explored the mechanisms of LF fibrosis at the molecular and cellular levels, the exact mechanism remains unknown. It is suggested that pathophysiologic stimuli such as mechanical stress, aging, obesity, and some diseases are the causative factors. Then, many cytokines and growth factors secreted by LF cells and its surrounding tissues play different roles in activating the fibrotic response. Here, we summarize the current status of detailed knowledge available regarding the causative factors, pathology, molecular and cellular mechanisms implicated in LF fibrosis and hypertrophy, also focusing on the possible avenues for anti-fibrotic strategies.
Collapse
Affiliation(s)
- Chao Sun
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Han Zhang
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xiang Wang
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Xinhui Liu
- Department of Spine Surgery, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Ye S, Kwon WK, Bae T, Kim S, Lee JB, Cho TH, Park JY, Kim K, Hur JK, Hur JW. CCN5 Reduces Ligamentum Flavum Hypertrophy by Modulating the TGF-β Pathway. J Orthop Res 2019; 37:2634-2644. [PMID: 31334871 PMCID: PMC6899892 DOI: 10.1002/jor.24425] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/17/2019] [Indexed: 02/04/2023]
Abstract
Ligamentum flavum hypertrophy (LFH) is the most important component of lumbar spinal canal stenosis. Although the pathophysiology of LFH has been extensively studied, no method has been proposed to prevent or treat it. Since the transforming growth factor-β (TGF-β) pathway is known to be critical in LFH pathology, we investigated whether LFH could be prevented by blocking or modulating the TGF-β mechanism. Human LF cells were used for the experiments. First, we created TGF-β receptor 1 (TGFBR1) knock out (KO) cells with CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 biotechnology and treated them with TGF-β1 to determine the effects of blocking the TGF-β pathway. Subsequently, we studied the effect of CCN5, which has recently been proposed to modulate the TGF-β pathway. To assess the predisposition toward fibrosis, α-smooth muscle actin (αSMA), fibronectin, collagen-1, collagen-3, and CCN2 were evaluated with quantitative real-time polymerase chain reaction, western blotting, and immunocytochemistry. The TGFBR1 KO LF cells were successfully constructed with high KO efficiency. In wild-type (WT) cells, treatment with TGF-β1 resulted in the overexpression of the messenger RNA (mRNA) of fibrosis-related factors. However, in KO cells, the responses to TGF-β1 stimulation were significantly lower. In addition, CCN5 and TGF-β1 co-treatment caused a notable reduction in mRNA expression levels compared with TGF-β1 stimulation only. The αSMA protein expression increased with TGF-β1 but decreased with CCN5 treatment. TGF-β1 induced LF cell transdifferentiation from fibroblasts to myofibroblasts. However, this cell transition dramatically decreased in the presence of CCN5. In conclusion, CCN5 could prevent LFH by modulating the TGF-β pathway. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. J Orthop Res 37:2634-2644, 2019.
Collapse
Affiliation(s)
| | - Woo-Keun Kwon
- Department of Neurosurgery, College of Medicine, Korea University Anam Hospital, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Taegeun Bae
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, Korea
| | - Sunghyun Kim
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, California
| | - Jang-Bo Lee
- Department of Neurosurgery, College of Medicine, Korea University Anam Hospital, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Tai-Hyoung Cho
- Department of Neurosurgery, College of Medicine, Korea University Anam Hospital, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Jung-Yul Park
- Department of Neurosurgery, College of Medicine, Korea University Anam Hospital, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea
| | - Kyoungmi Kim
- Department of Biomedical Sciences and Department of Physiology, College of Medicine, Korea University, Seoul, Korea
| | - Junho K Hur
- Department of Pathology, College of Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Korea
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul, Korea
| | - Junseok W Hur
- Department of Neurosurgery, College of Medicine, Korea University Anam Hospital, 73, Inchon-ro, Seongbuk-gu, Seoul, 02841, Korea
| |
Collapse
|
23
|
Xu Z, Li T, Li M, Yang L, Xiao R, Liu L, Chi X, Liu D. eRF3b-37 inhibits the TGF-β1-induced activation of hepatic stellate cells by regulating cell proliferation, G0/G1 arrest, apoptosis and migration. Int J Mol Med 2018; 42:3602-3612. [PMID: 30272252 DOI: 10.3892/ijmm.2018.3900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 09/20/2018] [Indexed: 11/05/2022] Open
Abstract
The therapeutic management of liver fibrosis remains an unresolved clinical problem. The activation of hepatic stellate cells (HSCs) serves a pivotal role in the formation of liver fibrosis. In our previous study, matrix‑assisted laser desorption/ionization time‑of‑flight mass spectrometry (MALDI‑TOF MS) was employed to identify potential serum markers for liver cirrhosis, such as eukaryotic peptide chain releasing factor 3b polypeptide (eRF3b‑37), which was initially confirmed by our group to serve a protective role in liver tissues in a C‑C motif chemokine ligand 4‑induced liver cirrhosis mouse model. Therefore, eRF3b‑37 was hypothesized to affect the activation state of HSCs, which was determined by the expression of pro‑fibrogenic associated factors in HSCs. In the present study, peptide synthesis technology was employed to elucidate the role of eRF3b‑37 in the expression of pro‑fibrogenic factors induced by transforming growth factor‑β1 (TGF‑β1) in LX‑2 cells that were treated with either control, TGF‑β1 and TGF‑β1+eRF3b‑37. 3‑(4,5‑Dimethyl‑2‑thiazolyl)‑2,5‑diphenyltetrazolium bromide and flow cytometric assays, and fluorescent microscope examinations were performed to evaluate the effects of eRF3b‑37 on proliferation viability, G0/G1 arrest, apoptosis and cell migration. The results of the present study indicated that eRF3b‑37 inhibited the activation of HSCs. The increased mRNA and protein expression of the pro‑fibrogenic factors collagen I, connective tissue growth factor and α‑smooth muscle actin (SMA) stimulated by TGF‑β1 were reduced by eRF3b‑37 via the following mechanisms: i) Inhibiting LX‑2 cell proliferation, leading to G0/G1 cell cycle arrest and inhibition of DNA synthesis by downregulating the mRNA expressions of Cyclin D1 and cyclin dependent kinase‑4, and upregulating the levels of P21; ii) increasing cell apoptosis by upregulating the mRNA level of B‑cell lymphoma-2 (Bcl‑2)‑associated X protein (Bax) and Fas, and downregulating the expression of Bcl‑2; and iii) reducing cell migration by downregulating the mRNA and protein expression of α‑SMA. In addition, eRF3b‑37 is thought to serve a role in HSCs by inhibiting TGF‑β signaling. Therefore, eRF3b‑37 may be a novel therapeutic agent for targeting HSCs for hepatic fibrosis.
Collapse
Affiliation(s)
- Zhengrong Xu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Tao Li
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Man Li
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Lei Yang
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Rudan Xiao
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Li Liu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xin Chi
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Dianwu Liu
- Department of Epidemiology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
24
|
Xu P, Zhang G, Hou S, Sha LG. MAPK8 mediates resistance to temozolomide and apoptosis of glioblastoma cells through MAPK signaling pathway. Biomed Pharmacother 2018; 106:1419-1427. [PMID: 30119215 DOI: 10.1016/j.biopha.2018.06.084] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/14/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE In this study, we aimed to evaluate the expression and functions of MAPK8 in temozolomide (TMZ) -resistant glioblastoma cells as well as to explore the mechanism of TMZ resistance in glioblastoma cells. METHODS Gene Expression Omnibus (GEO) database was used for identifying the differentially expressed genes (DEGs) in TMZ resistant samples. The functional partner genes of TMZ were screened out by Gene-drug interaction network (STITCH) and the glioblastoma-related genes were selected by gene search engine with evidence sentences (Digsee). The interactions among identified DEGs and glioblastoma-related genes were detected by Search Tool for the Retrieval of Interacting Genes (STRING). The dysregulated pathways were identified by Gene set enrichment analysis (GSEA). qRT-PCR was performed to detect the expression level of MAPK8 in glioblastoma cells. Western blot was used to detect the expressions of MAPK8 and MAPK signaling pathway-related proteins. MTT assay was utilized to measure the cell viability of TMZ sensitive and resistant cells. Colony formation assay was performed to detect the clone ability and flow cytometry (FCM) assay was applied to identify the apoptosis rate of TMZ resistant glioblastoma cells. RESULTS MAPK8 was one of the DEGs and was up-regulated in TMZ resistant glioblastoma cells. The MAPK signaling pathway was activated in TMZ resistant glioblastoma cells under the condition of over-expression of MAPK8. The inhibition of MAPK8 restrained the colony formation, inducing apoptosis of TMZ resistant glioblastoma cells and suppressed the MAPK signaling pathway. CONCLUSION MAPK8 promoted the resistance to TMZ, accelerated cell proliferation and inhibited the apoptosis of glioblastoma cells via activating MAPK signaling pathway.
Collapse
Affiliation(s)
- Peng Xu
- The Fourth Department of Geronotology, Jinan Military General Hospital, Jinan, 250031, Shandong, China
| | - Guofeng Zhang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Shaanxi, 710032, Xi'an, China
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Pudong, 201399, Shanghai, China.
| | - Long-Gui Sha
- Department of Neurosurgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, No. 2800 Gongwei Road, Pudong, 201399, Shanghai, China.
| |
Collapse
|
25
|
Papadopoulou-Marketou N, Kanaka-Gantenbein C, Marketos N, Chrousos GP, Papassotiriou I. Biomarkers of diabetic nephropathy: A 2017 update. Crit Rev Clin Lab Sci 2017; 54:326-342. [DOI: 10.1080/10408363.2017.1377682] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Nektaria Papadopoulou-Marketou
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
- Department of Endocrinology, Department of Medical and Health Sciences, Linkoping University, Linkoping, Sweden
| | - Christina Kanaka-Gantenbein
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
| | | | - George P. Chrousos
- Diabetes Centre of the Division of Endocrinology, Diabetes and Metabolism, First Department of Pediatrics, National and Kapodistrian University of Athens, “Aghia Sophia” Children’s Hospital, Athens, Greece
| | - Ioannis Papassotiriou
- Department of Clinical Biochemistry, “Aghia Sophia” Children’s Hospital, Athens, Greece
| |
Collapse
|
26
|
Saito T, Hara M, Kumamaru H, Kobayakawa K, Yokota K, Kijima K, Yoshizaki S, Harimaya K, Matsumoto Y, Kawaguchi K, Hayashida M, Inagaki Y, Shiba K, Nakashima Y, Okada S. Macrophage Infiltration Is a Causative Factor for Ligamentum Flavum Hypertrophy through the Activation of Collagen Production in Fibroblasts. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2831-2840. [PMID: 28935572 DOI: 10.1016/j.ajpath.2017.08.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 07/19/2017] [Accepted: 08/15/2017] [Indexed: 12/22/2022]
Abstract
Ligamentum flavum (LF) hypertrophy causes lumbar spinal canal stenosis, leading to leg pain and disability in activities of daily living in elderly individuals. Although previous studies have been performed on LF hypertrophy, its pathomechanisms have not been fully elucidated. In this study, we demonstrated that infiltrating macrophages were a causative factor for LF hypertrophy. Induction of macrophages into the mouse LF by applying a microinjury resulted in LF hypertrophy along with collagen accumulation and fibroblasts proliferation at the injured site, which were very similar to the characteristics observed in the severely hypertrophied LF of human. However, we found that macrophage depletion by injecting clodronate-containing liposomes counteracted LF hypertrophy even with microinjury. For identification of fibroblasts in the LF, we used collagen type I α2 linked to green fluorescent protein transgenic mice and selectively isolated green fluorescent protein-positive fibroblasts from the microinjured LF using laser microdissection. A quantitative RT-PCR on laser microdissection samples revealed that the gene expression of collagen markedly increased in the fibroblasts at the injured site with infiltrating macrophages compared with the uninjured location. These results suggested that macrophage infiltration was crucial for LF hypertrophy by stimulating collagen production in fibroblasts, providing better understanding of the pathophysiology of LF hypertrophy.
Collapse
Affiliation(s)
- Takeyuki Saito
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masamitsu Hara
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiromi Kumamaru
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazu Kobayakawa
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuya Yokota
- Department of Orthopaedic Surgery, Spinal Injuries Center, Fukuoka, Japan
| | - Ken Kijima
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shingo Yoshizaki
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsumi Harimaya
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Matsumoto
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kenichi Kawaguchi
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsumasa Hayashida
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Isehara, Japan; Department of Regenerative Medicine, School of Medicine, Tokai University, Isehara, Japan
| | - Keiichiro Shiba
- Department of Orthopaedic Surgery, Spinal Injuries Center, Fukuoka, Japan
| | - Yasuharu Nakashima
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Seiji Okada
- Department of Advanced Medical Initiatives, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
27
|
Sun X, Liu W, Cheng G, Qu X, Bi H, Cao Z, Yu Q. The influence of connective tissue growth factor on rabbit ligament injury repair. Bone Joint Res 2017; 6:399-404. [PMID: 28663339 PMCID: PMC5782798 DOI: 10.1302/2046-3758.67.bjr.2016-0255.r1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 02/28/2017] [Indexed: 01/13/2023] Open
Abstract
Objectives The injured anterior cruciate ligament (ACL) is thought to exhibit an impaired healing response, and attempts at surgical repair have not been successful. Connective tissue growth factor (CTGF) is reported to be associated with wound healing, probably through transforming growth factor beta 1 (TGF-β1). Methods A rabbit ACL injury model was used to study the effect of CTGF on ligament recovery. Quantitative real-time PCR (qRT-PCR) was performed for detection of changes in RNA levels of TGF-β1, type 1 collagen (COL1), type 2 collagen (COL2), SRY-related high mobility group-box gene9 (SOX9), tissue inhibitor of metalloproteinase-1 (TIMP-1) and matrix metallopeptidase 13 (MMP-13). Expression of related proteins was detected by Western blotting. Results The current study showed that CTGF could promote the recovery of an injured anterior cruciate ligament. It can upregulate mRNA and expression of TGF-β1, COL1, COL2, SOX9, and tissue inhibitor of TIMP-1, and downregulate mRNA and expression of MMP-13, suggesting that the curative effect of CTGF on injured rabbit ligaments is through regulation of these cellular factors. Conclusions This finding revealed the healing role of CTGF in injured tissues and provides new possibilities of treating injured tissues and wound healing by using CTGF. Cite this article: X. Sun, W. Liu, G. Cheng, X. Qu, H. Bi, Z. Cao, Q. Yu. The influence of connective tissue growth factor on rabbit ligament injury repair. Bone Joint Res 2017;6:399–404. DOI: 10.1302/2046-3758.67.BJR.2016-0255.R1.
Collapse
Affiliation(s)
- X Sun
- Yantaishan Hospital, Yantai, 264001, Shandong, China
| | - W Liu
- The Second Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - G Cheng
- Yantaishan Hospital, Yantai, 264001, Shandong, China
| | - X Qu
- Yantai Nursing School, Yantai, Shandong, 264000, China
| | - H Bi
- Yantaishan Hospital, Yantai, 264001, Shandong, China
| | - Z Cao
- Department of Orthopedics, Yantaishan Hospital, Yantai, 264001, Shandong, China
| | - Q Yu
- Operating Room, Yantaishan Hospital, Yantai, 264001, Shandong, China
| |
Collapse
|
28
|
Amudong A, Muheremu A, Abudourexiti T. Hypertrophy of the ligamentum flavum and expression of transforming growth factor beta. J Int Med Res 2017. [PMID: 28635357 PMCID: PMC5805210 DOI: 10.1177/0300060517711308] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Objective To explore the relationship between cellular apoptosis and hypertrophy of the
ligamentum flavum in the lumbar region. Methods Thirty patients with lumbar spinal stenosis were evaluated. Hypertrophy of
the ligamentum flavum was present in 15 patients and absent in 15.
Hematoxylin–eosin staining and transforming growth factor beta (TGF-β)
immunohistochemical testing were applied to compare these two groups. Results Derangement of fibrous alignment, fibrocartilage changes, and infiltration of
inflammatory cells were observed in the patients with hypertrophy of the
ligamentum flavum, while fibrous alignment was normal and few inflammatory
cells were observed in patients without hypertrophy. Immunohistochemical
studies showed positive expression of TGF-β in patients with hypertrophy,
while expression was negative in patients without hypertrophy. The
integrated optical density was 2.6556708 in the hypertrophy group and
23104671 in the normal controls. Conclusions Expression of TGF-β was closely related to hypertrophy of the ligamentum
flavum. Appropriate application of the TGF-β expression level can be used to
predict progression of hypertrophy of the ligamentum flavum.
Collapse
Affiliation(s)
- Aierken Amudong
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang Urumqi, China
| | - Aikeremujiang Muheremu
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang Urumqi, China
| | - Tuerhongjiang Abudourexiti
- Department of Spine Surgery, Sixth Affiliated Hospital of Xinjiang Medical University, Xinjiang Urumqi, China
| |
Collapse
|
29
|
Zhang W, Zheng J, Chen J, Huang L. The influence of connective tissue growth factor on rabbit ligament injury repair. Saudi Pharm J 2017; 25:498-503. [PMID: 28579882 PMCID: PMC5447422 DOI: 10.1016/j.jsps.2017.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Objectives The injured anterior cruciate ligament (ACL) is deemed to exhibit an impaired healing response and attempts at surgical repair have not been successful. Connective tissue growth factor (CTGF) is reported to be associated with wound healing, probably through transforming growth factor beta1 (TGF-β1). Methods A rabbit ACL injury model was used to study the effect of CTGF on ligament recovery. Quantitative real-time PCR was performed for detection of changes in RNA levels of TGF-β1, type 1 collagen (COL-I), type 2 collagen (COL-II), SRY-related high mobility group-box gene9 (Sox9), metalloproteinase-1 (TIMP-1) as well as matrix metallopeptidase 13 (MMP-13). And expression of related proteins was detected by western blotting. Results The current study showed that CTGF could promote the recovery of inured anterior cruciate ligament. It can up-regulate the mRNA and expression of TGF-β1, COL-I, COL-II, Sox9, as well as the tissue inhibitor of TIMP-1, and down-regulated the mRNA and expression of MMP-13, suggesting the curative effect of CTGF on injured rabbit ligament is through regulating these cellular factors. Conclusion This finding revealed the mechanism of CTFG’s healing role in injured tissues and provided new possibilities of treating injured tissues and wound healing by using CTFG.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Orthopedics, WenZhou Central Hospital, Wenzhou 325000, Zhejiang, China
| | - Junju Zheng
- Department of Orthopedics, WenZhou Central Hospital, Wenzhou 325000, Zhejiang, China
| | - Jiayu Chen
- Shaoxing University Medical School, Shaoxing 312000, Zhejiang, China
| | - Lipeng Huang
- Department of Orthopedics, WenZhou Central Hospital, Wenzhou 325000, Zhejiang, China
- Corresponding author.
| |
Collapse
|