1
|
Ganko R, Madhavan A, Hamouda W, Muthu S, Jain A, Yoon ST, El-Rozz H, Cyril D, Pabbruwe M, Tipper JL, Tavakoli J. Spinal implant wear particles: Generation, characterization, biological impacts, and future considerations. iScience 2025; 28:112193. [PMID: 40212584 PMCID: PMC11982499 DOI: 10.1016/j.isci.2025.112193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
The generation of wear debris from orthopedic implants is a known cause of implant failure, particularly in joint replacements. While much research has focused on wear particles from knee and hip implants, spinal implants, such as total disc replacements (TDRs), have received less attention despite their increasing clinical use. Spinal implants face unique biomechanical challenges, including a wider range of motion and higher loads, leading to complex tissue interactions. Studies reveal that TDR wear particles, though similar in size to those from knee implants, cause a stronger immune response, with more macrophages and giant cells found in the surrounding tissue. This may explain the high revision rates seen in spinal surgeries, with some interventions failing in over 30% of cases within 10 years. The younger population undergoing spinal surgery, combined with the productivity losses associated with implant failure, underscores the need for greater understanding. This review discusses recent research on the generation, characterization, and biological impacts of spinal implant wear debris. It draws on retrieval analysis, wear simulation, in vivo models, and a survey conducted with the AO Spine Knowledge Forum Degenerative to assess current clinical practices and highlight gaps in knowledge. Additionally, this critical review explores future strategies to reduce the biological impact of wear particles and improve the safety and longevity of spinal implants through better therapeutics and design innovations. By combining literature and clinical insights, this paper aims to guide future research in addressing the complexities of spinal implant wear and its biological consequences.
Collapse
Affiliation(s)
- Renata Ganko
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Aswini Madhavan
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Waeel Hamouda
- Department of Neurosurgery, Kasr Alainy Faculty of Medicine, Research, and Teaching Hospitals, Cairo University, Cairo, Egypt
- Department of Neurosurgery, Security Forces Hospital, Dammam, Saudi Arabia
| | - Sathish Muthu
- Department of Orthopaedics, Government Medical College, Karur, India
- Orthopaedic Research Group, Coimbatore, Tamil Nadu, India
- Department of Biotechnology, Karpagam Academy of Higher Education, Coimbatore, Tamil Nadu, India
| | - Amit Jain
- Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, MD, USA
| | - S. Tim Yoon
- Department of Orthopaedic Surgery, Emory University, Atlanta, GA, USA
| | - Hiba El-Rozz
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Divya Cyril
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
| | - Moreica Pabbruwe
- Centre for Implant Retrieval and Analysis, Royal Perth Hospital, Perth, WA, Australia
| | - Joanne L. Tipper
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
- School of Mechanical Engineering, University of Leeds, Leads, UK
| | - Javad Tavakoli
- School of Biomedical Engineering, Faculty of Eng and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| |
Collapse
|
2
|
Sukocheva OA, Neganova ME, Aleksandrova Y, Burcher JT, Chugunova E, Fan R, Tse E, Sethi G, Bishayee A, Liu J. Signaling controversy and future therapeutical perspectives of targeting sphingolipid network in cancer immune editing and resistance to tumor necrosis factor-α immunotherapy. Cell Commun Signal 2024; 22:251. [PMID: 38698424 PMCID: PMC11064425 DOI: 10.1186/s12964-024-01626-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 04/21/2024] [Indexed: 05/05/2024] Open
Abstract
Anticancer immune surveillance and immunotherapies trigger activation of cytotoxic cytokine signaling, including tumor necrosis factor-α (TNF-α) and TNF-related apoptosis-inducing ligand (TRAIL) pathways. The pro-inflammatory cytokine TNF-α may be secreted by stromal cells, tumor-associated macrophages, and by cancer cells, indicating a prominent role in the tumor microenvironment (TME). However, tumors manage to adapt, escape immune surveillance, and ultimately develop resistance to the cytotoxic effects of TNF-α. The mechanisms by which cancer cells evade host immunity is a central topic of current cancer research. Resistance to TNF-α is mediated by diverse molecular mechanisms, such as mutation or downregulation of TNF/TRAIL receptors, as well as activation of anti-apoptotic enzymes and transcription factors. TNF-α signaling is also mediated by sphingosine kinases (SphK1 and SphK2), which are responsible for synthesis of the growth-stimulating phospholipid, sphingosine-1-phosphate (S1P). Multiple studies have demonstrated the crucial role of S1P and its transmembrane receptors (S1PR) in both the regulation of inflammatory responses and progression of cancer. Considering that the SphK/S1P/S1PR axis mediates cancer resistance, this sphingolipid signaling pathway is of mechanistic significance when considering immunotherapy-resistant malignancies. However, the exact mechanism by which sphingolipids contribute to the evasion of immune surveillance and abrogation of TNF-α-induced apoptosis remains largely unclear. This study reviews mechanisms of TNF-α-resistance in cancer cells, with emphasis on the pro-survival and immunomodulatory effects of sphingolipids. Inhibition of SphK/S1P-linked pro-survival branch may facilitate reactivation of the pro-apoptotic TNF superfamily effects, although the role of SphK/S1P inhibitors in the regulation of the TME and lymphocyte trafficking should be thoroughly assessed in future studies.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia.
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center, Kazan Scientific Center, Russian Academy of Sciences, Kazan, 420088, Russian Federation
| | - Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Chernogolovka, 142432, Russian Federation
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center, Kazan Scientific Center, Russian Academy of Sciences, Kazan, 420088, Russian Federation
| | - Jack T Burcher
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Elena Chugunova
- Arbuzov Institute of Organic and Physical Chemistry, Federal Research Center, Kazan Scientific Center, Russian Academy of Sciences, Kazan, 420088, Russian Federation
| | - Ruitai Fan
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Edmund Tse
- Department of Hepatology, Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| | - Junqi Liu
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
3
|
Pan B, Zhang Z, Wu X, Xian G, Hu X, Gu M, Zheng L, Li X, Long L, Chen W, Sheng P. Macrophages-derived exosomes modulates wear particle-induced osteolysis via miR-3470b targeting TAB3/NF-κB signaling. Bioact Mater 2023; 26:181-193. [PMID: 36911207 PMCID: PMC9999169 DOI: 10.1016/j.bioactmat.2023.02.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 02/25/2023] [Accepted: 02/25/2023] [Indexed: 03/07/2023] Open
Abstract
Image 1.
Collapse
Key Words
- APL, Aseptic prothesis loosening
- Aseptic prothesis loosening
- Bglap, Osteocalcin
- CTSK, Cathepsin K
- Exosome
- Inflammatory osteolysis
- Macrophage
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NFATc-1, Nuclear factor of activated T-cells, cytoplasmic 1
- Non-coding RNA
- OB, Osteoblast
- OC, Osteoclast
- P-P65, phospho-P65
- P65, NF-κB signaling
- Runx2, Runt-related transcription factor 2
- TAB3, TGF-β-activated kinase 1 (MAP3K7) binding protein 3
- ncRNA, non-coding RNA
Collapse
Affiliation(s)
- Baiqi Pan
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Xiaoyu Wu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Guoyan Xian
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China.,Université de Paris, CNRS, INSERM, B3OA, Paris, France
| | - Xuantao Hu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Minghui Gu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Linli Zheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China.,Department of Spine Surgery, The first affiliated hospital of Sun Yat-sen University, China
| | - Lingli Long
- Research Center of Translational Medicine, The First Affiliated Hospital of Sun Yat-sen University, China
| | - Weishen Chen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, China
| |
Collapse
|
4
|
Maines LW, Green CL, Keller SN, Fitzpatrick LR, Smith CD. The Sphingosine Kinase 2 Inhibitor Opaganib Protects Against Acute Kidney Injury in Mice. Int J Nephrol Renovasc Dis 2022; 15:323-334. [PMID: 36420520 PMCID: PMC9677921 DOI: 10.2147/ijnrd.s386396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Acute kidney injury (AKI) is a common multifactorial adverse effect of surgery, circulatory obstruction, sepsis or drug/toxin exposure that often results in morbidity and mortality. Sphingolipid metabolism is a critical regulator of cell survival and pathologic inflammation processes involved in AKI. Opaganib (also known as ABC294640) is a first-in-class experimental drug targeting sphingolipid metabolism that reduces the production and activity of inflammatory cytokines and, therefore, may be effective to prevent and treat AKI. Methods Murine models of AKI were used to assess the in vivo efficacy of opaganib including ischemia-reperfusion (IR) injury induced by either transient bilateral occlusion of renal blood flow (a moderate model) or nephrectomy followed immediately by occlusion of the contralateral kidney (a severe model) and lipopolysaccharide (LPS)-induced sepsis. Biochemical and histologic assays were used to quantify the effects of oral opaganib treatment on renal damage in these models. Results Opaganib suppressed the elevations of creatinine and blood urea nitrogen (BUN), as well as granulocyte infiltration into the kidneys, of mice that experienced moderate IR from transient bilateral ligation. Opaganib also markedly decreased these parameters and completely prevented mortality in the severe renal IR model. Additionally, opaganib blunted the elevations of BUN, creatinine and inflammatory cytokines following exposure to LPS. Conclusion The data support the hypotheses that sphingolipid metabolism is a key mediator of renal inflammatory damage following IR injury and sepsis, and that this can be suppressed by opaganib. Because opaganib has already undergone clinical testing in other diseases (cancer and Covid-19), the present studies support conducting clinical trials with this drug with surgical or septic patients at risk for AKI.
Collapse
Affiliation(s)
- Lynn W Maines
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
| | | | | | | | - Charles D Smith
- Apogee Biotechnology Corporation, Hummelstown, PA, USA
- Correspondence: Charles D Smith, Apogee Biotechnology Corporation, 1214 Research Blvd, Suite 2015, Hummelstown, PA, 17036, USA, Email
| |
Collapse
|
5
|
Niu W, Chen Y, Wang L, Li J, Cui Z, Lv J, Yang F, Huo J, Zhang Z, Ju J. The combination of sodium alginate and chlorogenic acid enhances the therapeutic effect on ulcerative colitis by the regulation of inflammation and the intestinal flora. Food Funct 2022; 13:10710-10723. [PMID: 36173280 DOI: 10.1039/d2fo01619b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chlorogenic acid (CA) and sodium alginate (SA) each have good therapeutic effects on ulcerative colitis (UC) owing to their antioxidant and anti-inflammatory activity. This study aimed to investigate the effects of CA alone and in combination with SA on inflammatory cells and UC mice. In the Lipopolysaccharide (LPS)-induced RAW 264.7 inflammatory cell model, Nitric oxide (NO) and interleukin-6 (IL-6) levels were significantly lower after treatment with CA plus SA than with CA alone. In the DSS-induced UC mouse model, compared with CA alone, CA plus SA showed a better ability to alleviate weight loss, reduce the disease activity index (DAI), improve the colonic mucosa, reduce the expression of inflammatory factors in the serum and myeloperoxidase (MPO) in colonic tissue, increase superoxide dismutase (SOD) levels, protect the intestinal mucosa and regulate the abundance of Actinobacteriota, Lactobacillus, Bifidobacterium, Bacteroides, Subdoligranulum and Streptococcus. Thus, CA plus SA can improve the therapeutic efficacy of CA in UC by regulating inflammatory factors, oxidative stress, and the intestinal flora and by protecting ulcerative wounds. These findings broaden our understanding of the role of the combination of SA and CA in enhancing the effects of CA on UC and provide strategies for prevention and treatment.
Collapse
Affiliation(s)
- Wei Niu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yuxuan Chen
- School of Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ligui Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhao Cui
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jiajie Lv
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Fuyan Yang
- Anhui University of Chinese Medicine, Hefei, China
| | - Jiege Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jianming Ju
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Gu M, Pan B, Chen W, Xu H, Wu X, Hu X, Zheng L, Ye Y, Meng Q, Xian G, Zhang Z, Sheng P. SPHK Inhibitors and Zoledronic Acid Suppress Osteoclastogenesis and Wear Particle-Induced Osteolysis. Front Pharmacol 2022; 12:794429. [PMID: 35237148 PMCID: PMC8883393 DOI: 10.3389/fphar.2021.794429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Inflammatory osteolysis induced by wear particles is the major cause of prosthetic loosening after artificial joint replacement, and its prevention and treatment are difficult worldwide. Our previous study confirmed that sphingosine kinases (SPHKs) are important mediators regulating the wear particle-induced macrophage inflammatory response. However, it is unclear whether SPHKs can modulate chronic inflammation and alleviate osteolysis. Zoledronic acid (ZA), an imidazole-containing bisphosphonate, directly affects osteoclasts and prevents bone mineral-related diseases. However, the effects of SPHK inhibitors and ZA used to treat periprosthetic osteolysis are unknown. Methods: We applied tartrate-resistant acid phosphatase (TRAP) staining to evaluate bone destruction in the interface membranes of patients with aseptic loosening and a control group. A murine calvarial osteolysis model was used to examine the preventative effect of SPHK inhibitors and ZA on osteolysis. Micro-CT scanning, immunohistochemistry (IHC), and histomorphometric analysis were conducted to determine the variations in inflammatory osteolysis. The effects of different drug concentrations on cell viability were evaluated using the Cell Counting Kit-8 (CCK-8) assay. Real-time quantitative polymerase chain reaction (RT-qPCR) analysis was performed to confirm the reduced expression of osteoclast-specific genes after drug and titanium treatment. The osteoclast formation and functions of the drugs were analyzed using TRAP staining in vivo and in vitro. The effect of SPHKs/S1P-TRAF2-BECN1 signaling pathways was verified via RT-qPCR and tissue IHC. Results: In this study, we found that SPHK inhibitors (ABC294640 and FTY720) combined with ZA decreased the degree of inflammatory osteolysis in vivo. However, ABC294640 and ZA suppressed osteoclast differentiation and osteoclast-specific genes in vitro. SPHKs regulate the inflammatory osteolysis induced by wear particles by increasing the expression of SPHKs/S1P-TRAF2-BECN1. Conclusion: Our study revealed that wear particles could induce inflammatory osteolysis by upregulating SPHKs/S1P-TRAF2-BECN1 and SPHK inhibitors/ZA inhibit osteoclastogenesis in vitro and prevent inflammatory osteolysis in vivo, suggesting that SPHK inhibitors and ZA can be a new perspective and scientific basis for the prevention and treatment of prosthesis loosening.
Collapse
Affiliation(s)
- Minghui Gu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Baiqi Pan
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Weishen Chen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hai Xu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyu Wu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuantao Hu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Linli Zheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yongyu Ye
- Department of Orthopaedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Meng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Department of Orthopedics, Guizhou Orthopedics Hospital, Guiyang, China
| | - Guoyan Xian
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Université de Paris, CNRS, INSERM, B3OA, Paris, France
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
7
|
Jovanović JĐ, Antonijević M, El‐Emam AA, Marković Z. Comparative MD Study of Inhibitory Activity of Opaganib and Adamantane-Isothiourea Derivatives toward COVID-19 Main Protease M pro. ChemistrySelect 2021; 6:8603-8610. [PMID: 34909459 PMCID: PMC8662094 DOI: 10.1002/slct.202101898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/18/2021] [Indexed: 12/16/2022]
Abstract
In this study, the inhibitory potency of four adamantly- isothiourea derivatives (compounds 1 [4-bromobenzyl (Z)-N'-(adamantan-1-yl)-4-phenylpiperazine-1-carbothioimidate], 2 [3,5-bis(trifluoromethyl)benzyl (Z)-N'-(adamantan-1-yl)-4-phenylpiperazine-1-carbothioimidate], 3 [4-bromobenzyl (Z)-N-(adamantan-1-yl)morpholine-4-carbothioimidate] and 4 [3,5-bis(trifluoromethyl)benzyl (Z)-N-(adamantan-1-yl)morpholine-4-carbothioimidate]) was evaluated against SARS-CoV-2 targeted proteins. The investigated compounds 1-4 possess a similar structure to opaganib, which is used in studies like a potential drug for COVID-19 treatment. Since examined adamantly-isothiourea derivatives (1-4) shown broad-spectrum of antibacterial activity and significant in vitro cytotoxic effects against five human tumor cell lines and shown similarity in structure with opaganib, it was of interest to study their inhibitory potency toward some SARS-CoV-2 proteins such as SARS-CoV-2 main protease Mpro and mutation of SARS-CoV-2 Spike (S) Protein D614G. The inhibitory potency of studied compounds is examined using molecular docking and molecular dynamic simulations. The results of molecular docking simulations indicate compound 1 as the most prominent candidate of inhibition of SARS-CoV-2 main protease Mpro (▵Gbind=11.24 kcal/mol), while almost the same inhibition potency of all studied compounds is exhibited toward D614G. Regarding the results obtained by molecular dynamic simulations, compounds 1 and 4 possess similar inhibitory potency toward SARS-CoV-2 main protease Mpro as opaganib (▵Gbind ≈ 40 kcal/mol).
Collapse
Affiliation(s)
- Jelena Đorović Jovanović
- Department of ScienceInstitute for Information TechnologiesUniversity of Kragujevac, Jovana Cvijića bb34000Kragujevac, Republic ofSerbia
| | - Marko Antonijević
- Department of ScienceInstitute for Information TechnologiesUniversity of Kragujevac, Jovana Cvijića bb34000Kragujevac, Republic ofSerbia
| | - Ali A. El‐Emam
- Department of Medicinal ChemistryFaculty of PharmacyMansoura UniversityMansoura35516Egypt
| | - Zoran Marković
- Department of ScienceInstitute for Information TechnologiesUniversity of Kragujevac, Jovana Cvijića bb34000Kragujevac, Republic ofSerbia
| |
Collapse
|
8
|
Chen W, Xian G, Gu M, Pan B, Wu X, Ye Y, Zheng L, Zhang Z, Sheng P. Autophagy inhibitors 3-MA and LY294002 repress osteoclastogenesis and titanium particle-stimulated osteolysis. Biomater Sci 2021; 9:4922-4935. [PMID: 34052845 DOI: 10.1039/d1bm00691f] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Aseptic loosening caused by peri-implant osteolysis (PIO) is a common complication after joint replacement, and there is still no better treatment than revision surgery. The wear particle-induced inflammation response, especially subsequent osteoclastic bone resorption, is responsible for PIO. As the importance of wear particles in inducing autophagy in cells around the prosthesis in PIO has been discovered, this might be a central process underlying aseptic loosening. However, the role of autophagy induced by wear particles in osteoclastogenesis during PIO remains unclear. In this study, we investigated the role of autophagy in osteoclastogenesis and verified it in a mouse calvarial osteolysis model. We found that osteoclasts were increased in the interface membranes of patients with aseptic loosening. In vitro, knocking down the Atg5 gene or using autophagy inhibitors (3-MA, LY294002) to inhibit autophagy was found to repress osteoclastogenesis and decrease expression of the osteoclast-related genes TRAP, cathepsin K, and matrix metalloprotein 9 (MMP-9) with or without titanium (Ti) particles. In vivo, 3-MA and LY294002 repressed Ti particle-stimulated osteolysis and osteoclastogenesis and reduced expression of the pro-inflammatory factors TNF-α, IL-1β, and IL-6. Our results suggest that 3-MA and LY294002 might be the potential medicines to prevent and treat PIO and aseptic loosening.
Collapse
Affiliation(s)
- Weishen Chen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Guoyan Xian
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Minghui Gu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Baiqi Pan
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Xiaoyu Wu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yongyu Ye
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Linli Zheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China. and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
9
|
Pei G, Zyla J, He L, Moura‐Alves P, Steinle H, Saikali P, Lozza L, Nieuwenhuizen N, Weiner J, Mollenkopf H, Ellwanger K, Arnold C, Duan M, Dagil Y, Pashenkov M, Boneca IG, Kufer TA, Dorhoi A, Kaufmann SHE. Cellular stress promotes NOD1/2-dependent inflammation via the endogenous metabolite sphingosine-1-phosphate. EMBO J 2021; 40:e106272. [PMID: 33942347 PMCID: PMC8246065 DOI: 10.15252/embj.2020106272] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 03/25/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022] Open
Abstract
Cellular stress has been associated with inflammation, yet precise underlying mechanisms remain elusive. In this study, various unrelated stress inducers were employed to screen for sensors linking altered cellular homeostasis and inflammation. We identified the intracellular pattern recognition receptors NOD1/2, which sense bacterial peptidoglycans, as general stress sensors detecting perturbations of cellular homeostasis. NOD1/2 activation upon such perturbations required generation of the endogenous metabolite sphingosine-1-phosphate (S1P). Unlike peptidoglycan sensing via the leucine-rich repeats domain, cytosolic S1P directly bound to the nucleotide binding domains of NOD1/2, triggering NF-κB activation and inflammatory responses. In sum, we unveiled a hitherto unknown role of NOD1/2 in surveillance of cellular homeostasis through sensing of the cytosolic metabolite S1P. We propose S1P, an endogenous metabolite, as a novel NOD1/2 activator and NOD1/2 as molecular hubs integrating bacterial and metabolic cues.
Collapse
Affiliation(s)
- Gang Pei
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
- Present address:
Institute of ImmunologyFriedrich‐Loeffler‐InstitutGreifswald‐Insel RiemsGermany
| | - Joanna Zyla
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
- Department of Data Science and EngineeringSilesian University of TechnologyGliwicePoland
| | - Lichun He
- State Key Laboratory of Magnetic Resonance and Atomic Molecular PhysicsKey Laboratory of Magnetic Resonance in Biological SystemsNational Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Pedro Moura‐Alves
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
- Nuffield Department of MedicineLudwig Institute for Cancer ResearchUniversity of OxfordOxfordUK
| | - Heidrun Steinle
- Department of ImmunologyInstitute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - Philippe Saikali
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
| | - Laura Lozza
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
| | | | - January Weiner
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
| | | | - Kornelia Ellwanger
- Department of ImmunologyInstitute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - Christine Arnold
- Department of ImmunologyInstitute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - Mojie Duan
- State Key Laboratory of Magnetic Resonance and Atomic Molecular PhysicsKey Laboratory of Magnetic Resonance in Biological SystemsNational Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and TechnologyChinese Academy of SciencesWuhanChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yulia Dagil
- Institute of Immunology of the Federal Medical‐Biological Agency of RussiaMoscowRussia
| | - Mikhail Pashenkov
- Institute of Immunology of the Federal Medical‐Biological Agency of RussiaMoscowRussia
| | - Ivo Gomperts Boneca
- Institut PasteurDepartment of Microbiology, Biology and Genetics of the Bacterial Cell WallParisFrance
- CNRS UMR2001Integrative and Molecular MicrobiologyParisFrance
- INSERMÉquipe AVENIRParisFrance
| | - Thomas A Kufer
- Department of ImmunologyInstitute of Nutritional MedicineUniversity of HohenheimStuttgartGermany
| | - Anca Dorhoi
- Institute of ImmunologyFriedrich‐Loeffler‐InstitutGreifswald‐Insel RiemsGermany
- Faculty of Mathematics and Natural SciencesUniversity of GreifswaldGreifswaldGermany
| | - Stefan HE Kaufmann
- Department of ImmunologyMax Planck Institute for Infection BiologyBerlinGermany
- Hagler Institute for Advanced Study at Texas A&M UniversityCollege StationTXUSA
| |
Collapse
|
10
|
Sharma J, Parsai K, Raghuwanshi P, Ali SA, Tiwari V, Bhargava A, Mishra PK. Emerging role of mitochondria in airborne particulate matter-induced immunotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116242. [PMID: 33321436 DOI: 10.1016/j.envpol.2020.116242] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 05/05/2023]
Abstract
The immune system is one of the primary targets of airborne particulate matter. Recent evidence suggests that mitochondria lie at the center of particulate matter-induced immunotoxicity. Particulate matter can directly interact with mitochondrial components (proteins, lipids, and nucleic acids) and impairs the vital mitochondrial processes including redox mechanisms, fusion-fission, autophagy, and metabolic pathways. These disturbances impede different mitochondrial functions including ATP production, which acts as an important platform to regulate immunity and inflammatory responses. Moreover, the mitochondrial DNA released into the cytosol or in the extracellular milieu acts as a danger-associated molecular pattern and triggers the signaling pathways, involving cGAS-STING, TLR9, and NLRP3. In the present review, we discuss the emerging role of mitochondria in airborne particulate matter-induced immunotoxicity and its myriad biological consequences in health and disease.
Collapse
Affiliation(s)
- Jahnavi Sharma
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Kamakshi Parsai
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pragati Raghuwanshi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Sophiya Anjum Ali
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Vineeta Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India.
| |
Collapse
|
11
|
Xian G, Chen W, Gu M, Ye Y, Yang G, Lai W, Xiao Y, Zhao X, Zheng L, Pan B, Kang Y, Zhang Z, Sheng P. Titanium particles induce apoptosis by promoting autophagy in macrophages via the PI3K/Akt signaling pathway. J Biomed Mater Res A 2020; 108:1792-1805. [PMID: 32198815 DOI: 10.1002/jbm.a.36938] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 12/28/2022]
Abstract
Chronic inflammation and infection in the tissue surrounding implants after total joint replacement is closely associated with the innate immune response to surgical implants. Wear particles are known to increase apoptosis and impair the innate immunity in macrophages, which can cause immunosuppression around the implants. Excessive autophagy can induce apoptosis. However, the link between autophagy and apoptosis in macrophages during chronic inflammation and infection remains unknown. In this study, we investigated the autophagy and apoptosis induced by titanium particles in RAW264.7 macrophages, and in the interface membrane of patients with late-onset periprosthetic joint infection (PJI). We found that titanium particles stimulated autophagy and apoptosis in macrophages. Inhibition of autophagy significantly reduced titanium particle-induced apoptosis in macrophages, which may be related to the PI3K/Akt signaling pathway. The secretion of inflammatory factors, such as IL-1β, IL-6, and TNF-α, decreased after inhibition of autophagy in titanium particle-stimulated macrophages, which may be caused by immune dysfunction due to titanium particle-induced autophagy and apoptosis in macrophages. Furthermore, our in vivo mouse calvarial model also showed that autophagy inhibitors lowered the rate of cell apoptosis. Our findings indicate that wear particle-induced apoptosis may be caused by enhanced autophagy in macrophages, which could potentially impair the local innate immunity in periprosthetic tissues and could be a risk factor for PJI. Based on these results, autophagy modulators may act as a new therapeutic option for PJI.
Collapse
Affiliation(s)
- Guoyan Xian
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weishen Chen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Minghui Gu
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yongyu Ye
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guangpu Yang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Weiming Lai
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yinbo Xiao
- Centre for the Cellular Microenvironment, Institute of Molecular, Cell & Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Xiaoyi Zhao
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Linli Zheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Baiqi Pan
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yunze Kang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ziji Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Puyi Sheng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
12
|
Yang X, Guo JL, Han J, Si RJ, Liu PP, Zhang ZR, Wang AM, Zhang J. Chitosan hydrogel encapsulated with LL-37 peptide promotes deep tissue injury healing in a mouse model. Mil Med Res 2020; 7:20. [PMID: 32321591 PMCID: PMC7175584 DOI: 10.1186/s40779-020-00249-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 04/08/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND LL-37 peptide is a member of the human cathelicidin family, and has been shown to promote the healing of pressure ulcers. However, the low stability of this peptide within the wound environment limits its clinical use. Chitosan (CS) hydrogel is commonly used as a base material for wound dressing material. METHODS CS hydrogel (2.5% w/v) was encapsulated with LL-37. Cytotoxicity of the product was examined in cultured NIH3T3 fibroblasts. Effects on immune response was examined by measuring tumor necrosis factor-α (TNF-α) release from RAW 264.7 macrophages upon exposure to lipopolysaccharides. Antibacterial activity was assessed using Staphylococcus aureus. Potential effect on pressure ulcers was examined using a mouse model. Briefly, adult male C57BL/6 mice were subjected to skin pressure using magnets under a 12/12 h schedule for 21 days. Mice were randomized to receive naked LL-37 (20 μg), chitosan gel containing 20-μg LL-37 (LL-37/CS hydrogel) or hydrogel alone under the ulcer bed (n = 6). A group of mice receiving no intervention was also included as a control. RESULTS LL-37/CS hydrogel did not affect NIH3T3 cell viability. At a concentration of 1-5 μg/ml, LL-37/CS inhibited TNF-α release from macrophage. At 5 μg/ml, LL-37/CS inhibited the growth of Staphylococcus aureus. The area of the pressure ulcers was significantly lower in mice receiving LL-37/CS hydrogel in comparison to all other 3 groups on days 11 (84.24% ± 0.25%), 13 (56.22% ± 3.91%) and 15 (48.12% ± 0.28%). Histological examination on days 15 and 21 showed increased epithelial thickness and density of newly-formed capillary with naked LL-37 and more so with LL-37/CS. The expression of key macromolecules in the process of angiogenesis (i.e., hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor-A (VEGF-A)) in wound tissue was increased at both the mRNA and protein levels. CONCLUSION Chitosan hydrogel encapsulated with LL-37 is biocompatible and could promote the healing of pressure ulcers.
Collapse
Affiliation(s)
- Xu Yang
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Jing-Lin Guo
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Jing Han
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Rui-Juan Si
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Pan-Pan Liu
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Zi-Rui Zhang
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Ai-Min Wang
- School of Nursing, Qingdao University, Qingdao, 266021, China
| | - Ju Zhang
- School of Nursing, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
13
|
The Immunosuppressant Fingolimod (FTY720) for the Treatment of Mechanical Force-Induced Abnormal Scars. J Immunol Res 2020; 2020:7057195. [PMID: 32377536 PMCID: PMC7199562 DOI: 10.1155/2020/7057195] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 01/29/2023] Open
Abstract
Aim Abnormal scars such as hypertrophic scars (HSs) and keloids are excessively growing scars that exhibit chronic inflammation and capillary vasculogenesis. The lipid mediator sphingosine-1-phosphate (S1P) is important in inflammatory cell recruitment and angiogenesis. Fingolimod (FTY720) is an analog of S1P and thus functionally antagonizes S1P receptors and inhibits the enzyme that produces S1P. We examined the effects of topical FTY720 injections on mechanical force-induced HS progression. Methods Mechanical force-induced HSs were generated in C57BL6/J mice by suturing a dorsal incision and applying a stretching device on Days 6, 8, 10, and 12. On Days 8, 10, and 12, intracutaneous FTY720 (10 μM) or control vehicle injections were performed. On Day 14, scar tissues and blood were procured and subjected to histology and flow cytometry. Results Flow cytometry showed that FTY720 decreased the frequencies of macrophages with M2 predominance in the scars but had no effect on total, CD4+, or CD8a+ T cell frequencies. FTY720 also decreased the vascular endothelial cell frequencies in the scar along with the microvessels, as determined by immunohistochemistry. Compared to the vehicles, FTY720 treatment significantly reduced the gross scar area and the cross-sectional scar area on histology. On the other hand, FTY720 tended to reduce white blood cells and significantly reduced the lymphocyte frequencies in the blood. Conclusion Topical FTY720 induces M2 predominance and impairs angiogenesis. Therefore, its local immunosuppressive mechanisms differ from those of conventional immunosuppressive agents. Topical FTY720 can be a novel therapeutic option for abnormal scars that are difficult to control with corticosteroids. Its lymphocytopenic effects may be limited by careful optimization of the treatment regimen.
Collapse
|
14
|
Liu J, Jiang B. Sphk1 promotes ulcerative colitis via activating JAK2/STAT3 signaling pathway. Hum Cell 2019; 33:57-66. [PMID: 31606874 DOI: 10.1007/s13577-019-00283-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/15/2019] [Indexed: 12/11/2022]
Abstract
Ulcerative colitis (UC) is a chronic non-specific inflammatory disease of the colon and rectum. The cause of ulcerative colitis is still unclear, although there may be a hereditary factor. SphK1 has been reported to exhibit an inhibitory effect on the occurrence and development of inflammation; however, the association between SphK1 and the progression of UC remains unclear. The aim of the present study was to investigate the effect of Sphk1 on the progression of UC. The proliferation of RAW264.7 cells was determined using a Cell Counting Kit-8 assay and apoptosis was measured using flow cytometry. The levels of pro-inflammatory cytokines secreted by RAW264.7 cells were investigated using ELISA kits and the protein expression levels in RAW264.7 cells were examined by western blotting. A dextran sulfate sodium (DSS)-induced mouse model was established to investigate the effect of SphK1 on the progression of UC in vivo. Overexpression of Sphk1 significantly increased the proliferation and inhibited the apoptosis of RAW264.7 cells. Additionally, overexpression of Sphk1 increased the secretion of pro-inflammatory cytokines and activated the JAK2/STAT3 signaling pathway in RAW264.7 cells, and JSI-124 partially suppressed these effects. Furthermore, SphK1-small interfering RNA or JSI-124 partially rescued lipopolysaccharide-induced proliferation and pro-inflammatory effects on RAW264.7 cells. The SphK1 inhibitor (PF-543) had an inhibitory effect on DSS-induced UC mice. Sphk1 had significant pro-inflammatory effects on the progression of UC, and may thus be a potential novel therapeutic target for the treatment of UC.
Collapse
Affiliation(s)
- Jiawen Liu
- Department of Gastroenterology, Beijing TsingHua Changgung Hospital, No. 168, LiTang Road, Beijing, 102218, China
| | - Bo Jiang
- Department of Gastroenterology, Beijing TsingHua Changgung Hospital, No. 168, LiTang Road, Beijing, 102218, China.
| |
Collapse
|