1
|
Tu H, Li YL. Inflammation balance in skeletal muscle damage and repair. Front Immunol 2023; 14:1133355. [PMID: 36776867 PMCID: PMC9909416 DOI: 10.3389/fimmu.2023.1133355] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/12/2023] [Indexed: 01/27/2023] Open
Abstract
Responding to tissue injury, skeletal muscles undergo the tissue destruction and reconstruction accompanied with inflammation. The immune system recognizes the molecules released from or exposed on the damaged tissue. In the local minor tissue damage, tissue-resident macrophages sequester pro-inflammatory debris to prevent initiation of inflammation. In most cases of the skeletal muscle injury, however, a cascade of inflammation will be initiated through activation of local macrophages and mast cells and recruitment of immune cells from blood circulation to the injured site by recongnization of damage-associated molecular patterns (DAMPs) and activated complement system. During the inflammation, macrophages and neutrophils scavenge the tissue debris to release inflammatory cytokines and the latter stimulates myoblast fusion and vascularization to promote injured muscle repair. On the other hand, an abundance of released inflammatory cytokines and chemokines causes the profound hyper-inflammation and mobilization of immune cells to trigger a vicious cycle and lead to the cytokine storm. The cytokine storm results in the elevation of cytolytic and cytotoxic molecules and reactive oxygen species (ROS) in the damaged muscle to aggravates the tissue injury, including the healthy bystander tissue. Severe inflammation in the skeletal muscle can lead to rhabdomyolysis and cause sepsis-like systemic inflammation response syndrome (SIRS) and remote organ damage. Therefore, understanding more details on the involvement of inflammatory factors and immune cells in the skeletal muscle damage and repair can provide the new precise therapeutic strategies, including attenuation of the muscle damage and promotion of the muscle repair.
Collapse
|
2
|
Mohammad G, Kowluru RA. Involvement of High Mobility Group Box 1 Protein in Optic Nerve Damage in Diabetes. Eye Brain 2022; 14:59-69. [PMID: 35586662 PMCID: PMC9109986 DOI: 10.2147/eb.s352730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/16/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Diabetic patients routinely have high levels of high mobility group box 1 (HMGB1) protein in their plasma, vitreous and ocular membranes, which is strongly correlated with subclinical chronic inflammation in the eye. Our previous work has suggested that high HMGB1 in diabetes plays a role in retinal inflammation and angiogenesis, but its role in the optic nerve damage is unclear. Therefore, our goal is to examine the role of HMGB1 in optic nerve damage in diabetes. Methods Gene expression of HMGB1 was quantified in the optic nerve from streptozotocin-induced diabetic mice by qRT-PCR, and their protein expressions by Western blot analysis and immunofluorescence staining. Using immunohistochemical technique, expression of reactive astrogliosis (indicator of neuroinflammation) and nerve demyelination/damage were determined by quantifying glial fibrillary acid protein (GFAP) and myelin basic protein (MBP), respectively. The role of HMGB1 in the optic nerve damage and alteration visual pathways was confirmed in mice receiving glycyrrhizin, a HMGB1 inhibitor. Similar parameters were measured in the optic nerve from human donors with diabetes. Results Compared to normal mice, diabetic mice exhibited increased levels of HMGB1, higher GFAP expression, and decreased MBP in the optic nerve. Double immunofluorescence microscopy revealed that diabetes induced increased HMGB1 immunoreactivities were significantly colocalized with GFAP in the optic nerve. Glycyrrhizin supplementation effectively reduced HMGB1 and maintained normal axonal myelination and visual conduction. Results from mice optic nerve confirmed the results obtained from human donors with diabetes. Discussions Thus, diabetes-induced HMGB1 upregulation promotes optic nerve demyelination and inflammation. The regulation of HMGB1 activation has potential to protect optic nerve damage and the abnormalities of visual pathways in diabetic patients.
Collapse
Affiliation(s)
- Ghulam Mohammad
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, 48201, USA
- Correspondence: Ghulam Mohammad, Tel +1 313-577-0744, Email
| | - Renu A Kowluru
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University, Detroit, MI, 48201, USA
- Kresge Eye Institute, Wayne State University, Detroit, MI, 48201, USA
| |
Collapse
|
3
|
Mahaling B, Low SWY, Beck M, Kumar D, Ahmed S, Connor TB, Ahmad B, Chaurasia SS. Damage-Associated Molecular Patterns (DAMPs) in Retinal Disorders. Int J Mol Sci 2022; 23:ijms23052591. [PMID: 35269741 PMCID: PMC8910759 DOI: 10.3390/ijms23052591] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/22/2022] [Accepted: 02/25/2022] [Indexed: 12/13/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are endogenous danger molecules released from the extracellular and intracellular space of damaged tissue or dead cells. Recent evidence indicates that DAMPs are associated with the sterile inflammation caused by aging, increased ocular pressure, high glucose, oxidative stress, ischemia, mechanical trauma, stress, or environmental conditions, in retinal diseases. DAMPs activate the innate immune system, suggesting their role to be protective, but may promote pathological inflammation and angiogenesis in response to the chronic insult or injury. DAMPs are recognized by specialized innate immune receptors, such as receptors for advanced glycation end products (RAGE), toll-like receptors (TLRs) and the NOD-like receptor family (NLRs), and purine receptor 7 (P2X7), in systemic diseases. However, studies describing the role of DAMPs in retinal disorders are meager. Here, we extensively reviewed the role of DAMPs in retinal disorders, including endophthalmitis, uveitis, glaucoma, ocular cancer, ischemic retinopathies, diabetic retinopathy, age-related macular degeneration, rhegmatogenous retinal detachment, proliferative vitreoretinopathy, and inherited retinal disorders. Finally, we discussed DAMPs as biomarkers, therapeutic targets, and therapeutic agents for retinal disorders.
Collapse
Affiliation(s)
- Binapani Mahaling
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
| | - Shermaine W. Y. Low
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
| | - Molly Beck
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
| | - Devesh Kumar
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
| | - Simrah Ahmed
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
| | - Thomas B. Connor
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
- Vitreoretinal Surgery, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Baseer Ahmad
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
- Vitreoretinal Surgery, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shyam S. Chaurasia
- Ocular Immunology and Angiogenesis Lab, Department of Ophthalmology and Visual Sciences, Froedtert and MCW Eye Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (B.M.); (S.W.Y.L.); (M.B.); (D.K.); (S.A.); (T.B.C.); (B.A.)
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Correspondence: ; Tel.: +1-414-955-2050
| |
Collapse
|
4
|
Ibrahim KG, Mukonowenzou NC, Usman D, Adeshina KA, Erlwanger KH. The potential of Artemisia species for use as broad-spectrum agents in the management of metabolic syndrome: a review. Arch Physiol Biochem 2021; 129:752-770. [PMID: 33569991 DOI: 10.1080/13813455.2021.1871761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Although the prevalence of metabolic syndrome (MetS), a cluster of cardiometabolic risk factors that predispose to the development of type 2 diabetes mellitus and cardiovascular diseases, is increasing globally, there is no broad-spectrum agent for its holistic treatment. Natural plant-derived products with a wide spectrum of biological activities are currently being explored as alternatives in the management of diseases. Artemisia species are a heterozygous group of plants of the Compositae family that possess several health benefits. Here we highlight their antidiabetic, anti-obesity, anti-hyperlipidaemic, hepatoprotective and cardioprotective properties among others. These activities have been linked to the presence of phytochemicals that act on several molecular targets to exert their effects and the species of Artemisia are considered to be relatively safe. Artemisia species offer significant anti-MetS activity and thus are strong therapeutic candidates for the effective management of MetS.
Collapse
Affiliation(s)
- Kasimu Ghandi Ibrahim
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Nyasha Charity Mukonowenzou
- Department of Anatomy and Physiology, Faculty of Medicine, National University of Science and Technology, Bulawayo, Zimbabwe
| | - Dawoud Usman
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Kehinde Ahmad Adeshina
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, Sokoto, Nigeria
- Centre for Advanced Medical Research and Training, Usmanu Danfodiyo University, Sokoto, Nigeria
| | - Kennedy Honey Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
5
|
Hou Y, Xin M, Li Q, Wu X. Glycyrrhizin micelle as a genistein nanocarrier: Synergistically promoting corneal epithelial wound healing through blockage of the HMGB1 signaling pathway in diabetic mice. Exp Eye Res 2021; 204:108454. [PMID: 33497689 DOI: 10.1016/j.exer.2021.108454] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/02/2021] [Accepted: 01/13/2021] [Indexed: 12/28/2022]
Abstract
The purpose of this study was to explore the feasibility of targeting the HMGB1 signaling pathway to treat diabetic keratopathy with a dipotassium glycyrrhizinate-based micelle ophthalmic solution encapsulating genistein (DG-Gen), and to evaluate whether these dipotassium glycyrrhizinate (DG) micelles could synergistically enhance the therapeutic effect of encapsulated genistein (Gen). An optimized DG-Gen ophthalmic solution was fabricated with a Gen/DG weight of ratio 1:15, and this formulation featured an encapsulation efficiency of 98.96 ± 0.82%, and an average particle size of 29.50 ± 2.05 nm. The DG-Gen ophthalmic solution was observed to have good in vivo ocular tolerance and excellent in vivo corneal permeation, and to remarkably improve in vitro antioxidant activity. Ocular topical application of the DG-Gen ophthalmic solution significantly prompted corneal re-epithelialization and nerve regeneration in diabetic mice, and this efficacy might be due to the inhibition of HMGB1 signaling through down-regulation of HMGB1 and its receptors RAGE and TLR4, as well as inflammatory factor interleukin (IL)-6 and IL-1β. In conclusion, these data showed that HMGB1 signaling is a potential regulation target for the treatment of diabetic keratopathy, and novel DG-micelle formulation encapsulating active agents such as Gen could synergistically cause blockage of HMGB1 signaling to prompt diabetic corneal and nerve wound healing.
Collapse
Affiliation(s)
- Yuzhen Hou
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Meng Xin
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China; Department of Ophthalmology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, 264100, China
| | - Qiqi Li
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Xianggen Wu
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China; Key Laboratory of Pharmaceutical Research for Metabolic Diseases, Qingdao University of Science and Technology, Qingdao, China.
| |
Collapse
|
6
|
Liu J, Wei E, Wei J, Zhou W, Webster KA, Zhang B, Li D, Zhang G, Wei Y, Long Y, Qi X, Zhang Q, Xu D. MiR-126-HMGB1-HIF-1 Axis Regulates Endothelial Cell Inflammation during Exposure to Hypoxia-Acidosis. DISEASE MARKERS 2021; 2021:4933194. [PMID: 34970357 PMCID: PMC8714334 DOI: 10.1155/2021/4933194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/20/2021] [Indexed: 02/05/2023]
Abstract
Crosstalk between molecular regulators miR-126, hypoxia-inducible factor 1-alpha (HIF-1-α), and high-mobility group box-1 (HMGB1) contributes to the regulation of inflammation and angiogenesis in multiple physiological and pathophysiological settings. Here, we present evidence of an overriding role for miR-126 in the regulation of HMGB1 and its downstream proinflammatory effectors in endothelial cells subjected to hypoxia with concurrent acidosis (H/A). Methods. Primary mouse endothelial cells (PMEC) were exposed to hypoxia or H/A to simulate short or chronic low-flow ischemia, respectively. RT-qPCR quantified mRNA transcripts, and proteins were measured by western blot. ROS were quantified by fluorogenic ELISA and luciferase reporter assays employed to confirm an active miR-126 target in the HMGB1 3'UTR. Results. Enhanced expression of miR-126 in PMECs cultured under neutral hypoxia was suppressed under H/A, whereas the HMGB1 expression increased sequentially under both conditions. Enhanced expression of HMGB1 and downstream inflammation markers was blocked by the premiR-126 overexpression and optimized by antagomiR. Compared with neutral hypoxia, H/A suppressed the HIF-1α expression independently of miR-126. The results show that HMGB1 and downstream effectors are optimally induced by H/A relative to neutral hypoxia via crosstalk between hypoxia signaling, miR-126, and HIF-1α, whereas B-cell lymphoma 2(Bcl2), a HIF-1α, and miR-126 regulated gene expressed optimally under neutral hypoxia. Conclusion. Inflammatory responses of ECs to H/A are dynamically regulated by the combined actions of hypoxia, miR-126, and HIF-1α on the master regulator HMGB1. The findings may be relevant to vascular diseases including atherosclerotic occlusion and interiors of plaque where coexisting hypoxia and acidosis promote inflammation as a defining etiology.
Collapse
Affiliation(s)
- Jinxue Liu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Eileen Wei
- Gulliver High School, Miami, FL 33156, USA
| | - Jianqin Wei
- Department of Medicine Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Wei Zhou
- Department of Ophthalmology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Keith A. Webster
- Integene International, LLC, Miami, FL 33137, USA
- Cullen Eye Institute, Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA
- Everglades Biopharma, LLC, Houston, TX 77030, USA
| | - Bin Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Dong Li
- Department of Intensive Care Unit and Clinical Experimental Center, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Gaoxing Zhang
- Department of Cardiology, Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-Sen University, Jiangmen 529030, China
| | - Yidong Wei
- Department of Surgery, Youjiang Medical University for Nationalities, Chengxiang Rd, Baise, Guangxi 533000, China
| | - Yusheng Long
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Second School of Clinical Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiuyu Qi
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
- Department of Cardiology, Guangdong Cardiovascular Institute and Shantou University Medical College, Shantou 515041, China
| | - Qianhuan Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou 510080, China
| | - Dingli Xu
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
7
|
Zhou Y, Wang T, Wang Y, Meng F, Ying M, Han R, Hao P, Wang L, Li X. Blockade of extracellular high-mobility group box 1 attenuates inflammation-mediated damage and haze grade in mice with corneal wounds. Int Immunopharmacol 2020; 83:106468. [PMID: 32279044 DOI: 10.1016/j.intimp.2020.106468] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/16/2020] [Accepted: 03/30/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE To investigate the expression of extracellular high mobility group box 1 (HMGB1) and the effect of its inhibitor glycyrrhizin (GL) in corneal wound healing. METHODS We treated C57BL/6J mice with GL or PBS before and after establishing a corneal injury model. Fluorescein staining, Ki-67 expression, haze grade, and haematoxylin/eosin (H&E) staining were used to assess treatment efficacy. The expression of HMGB1, NF-κB-p65, the NLRP3 inflammasome, IL-1β, CCL2, CXCL2, TGF-β1, α-SMA, fibronectin, and collagen III and neutrophil influx were examined by immunohistochemical staining, western blot, and RT-qPCR at various time points after corneal injury. RESULTS After corneal injury, HMGB1 transferred from the nucleus to the cytoplasm and was passively released or actively secreted into the corneal stroma from epithelial cells and inflammatory cells; however, this increase was attenuated by GL treatment. Furthermore, GL indirectly attenuated the expression of IL-1β by directly inhibiting extracellular HMGB1 functions, which activated the NF-κB-p65/NLRP3/IL-1β signalling pathway. Moreover, application of GL alleviated the neutrophil infiltration that delays wound healing, accompanied by the downregulation of expression of the chemokines CCL2 and CXCL2. More interestingly, application of GL reduced the degree of haze grade through inactivating extracellular HMGB1 functions that induced TGF-β1 release and myofibroblast differentiation. In addition, fluorescein and H&E staining and Ki-67 levels suggest that GL promotes regeneration of corneal epithelium. CONCLUSIONS After corneal injury, extracellular HMGB1 can be an essential driver to trigger a neutrophil- and cytokine-mediated inflammatory injury amplification loop. The application of GL promotes the cornea to restore transparency and integrity, which may be related to the attenuation of extracellular HMGB1 levels and function.
Collapse
Affiliation(s)
- Yongying Zhou
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China
| | - Ting Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Yuchuan Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China
| | - Fanlan Meng
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ming Ying
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China
| | - Ruifang Han
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China
| | - Peng Hao
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China
| | - Liming Wang
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China
| | - Xuan Li
- Clinical College of Ophthalmology, Tianjin Medical University, Tianjin, China; Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin, China; Nankai University Eye Hospital, Tianjin, China.
| |
Collapse
|
8
|
Pei X, Meng S, Gou C, Du Q. [Expression of high mobility group protein B1 in periodontal tissues and its association with hepatic lipid metabolism in diabetic rats with periodontitis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:6-12. [PMID: 32376562 DOI: 10.12122/j.issn.1673-4254.2020.01.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE To investigate the expression of high mobility group box-1 protein (HMGB1) and its downstream products, receptor for advanced glycation end-products (RAGE) and tumor necrosis factor-α (TNF-α), in periodontal tissues of diabetic rats with periodontitis, and explore the association of HMGB1 with hepatic lipid metabolism. METHODS Immunohistochemical staining was used to detect the expression of HMGB1, RAGE and TNF-α in the periodontal tissues in rat models of diabetes mellitus (DM), periodontitis (CP), and diabetic periodontitis (DM + CP). The serum levels of the indicators of lipid metabolism and biochemical indexes of liver damage were detected by spectroscopy. RESULTS The expressions of HMGB1 and RAGE in the periodontal tissues were significantly higher in DM group than in the control group, but the expression of TNF-α showed no significant difference among the groups. In CP group, the expressions of HMGB1 and TNF-α were significantly higher than those in the control group, and the expression of RAGE was comparable with that in the control group but significantly lower than that in DM and DM+CP group. The expressions of HMGB1, RAGE and TNF-α were all significantly higher in DM+CP group than in the control group. Compared with the control rats, the rats in DM, CP, DM+CP group all showed abnormal hepatic lipid metabolism with significantly elevated serum ALT levels. CONCLUSIONS HMGB1 and RAGE participate in the inflammation of the periodontal tissues in diabetic rats. Diabetes leads to elevated expression of HMGB1 in the periodontal tissues. Both periodontitis and hyperglycemia contribute to liver metabolic dysfunction. HMGB1- RAGE provides clues in the study of signaling pathways underlying the mutual susceptibility of diabetes and periodontitis.
Collapse
Affiliation(s)
- Xinfo Pei
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,Department of Stomatology, Xinhua Hospital Affiliated To Shanghai Jiaotong University, Shanghai 200092, China
| | - Shu Meng
- Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.,State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Chengdu 610041, China
| | - Ce Gou
- West China School of Stomatology, Sichuan University, Chengdu 610041, China
| | - Qin Du
- Department of Stomatology, Sichuan Academy of Medical Science and Sichuan Provincial People's Hospital, Chengdu 610072, China.,School of Medicine, University of Electronic Science and Technology, Chengdu 610054, China
| |
Collapse
|