1
|
Kiełbowski K, Stańska W, Bakinowska E, Rusiński M, Pawlik A. The Role of Alarmins in the Pathogenesis of Rheumatoid Arthritis, Osteoarthritis, and Psoriasis. Curr Issues Mol Biol 2024; 46:3640-3675. [PMID: 38666958 PMCID: PMC11049642 DOI: 10.3390/cimb46040228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Alarmins are immune-activating factors released after cellular injury or death. By secreting alarmins, cells can interact with immune cells and induce a variety of inflammatory responses. The broad family of alarmins involves several members, such as high-mobility group box 1, S100 proteins, interleukin-33, and heat shock proteins, among others. Studies have found that the concentrations and expression profiles of alarmins are altered in immune-mediated diseases. Furthermore, they are involved in the pathogenesis of inflammatory conditions. The aim of this narrative review is to present the current evidence on the role of alarmins in rheumatoid arthritis, osteoarthritis, and psoriasis. We discuss their potential involvement in mechanisms underlying the progression of these diseases and whether they could become therapeutic targets. Moreover, we summarize the impact of pharmacological agents used in the treatment of these diseases on the expression of alarmins.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Wiktoria Stańska
- Department of Medical Biology, Medical University of Warsaw, 00-575 Warsaw, Poland;
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Marcin Rusiński
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland; (K.K.); (E.B.); (M.R.)
| |
Collapse
|
2
|
Qin Q, Li Z, Liu R, Liu S, Guo M, Zhang M, Wu H, Huang L. Effects of resveratrol on HIF-1α/VEGF pathway and apoptosis in vitrified duck ovary transplantation. Theriogenology 2023; 210:84-93. [PMID: 37481978 DOI: 10.1016/j.theriogenology.2023.06.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023]
Abstract
Preservation of ovarian tissues is an effective way to ensure genetic diversity of susceptible natural bird populations that are in danger of extinction. We examined whether the addition of the plant phenol resveratrol to vitrification solutions ameliorates the damaging effects of tissue hypoxia and reperfusion injury when the tissues are transplanted. Duck ovary tissues were frozen in the presence of varying concentrations of resveratrol in cryopreservation solutions and then transplanted under the renal capsules of 2-day-old Shelducks. Samples of the transplanted tissues were examined on days 3- and 9- post transplantation for activation of hypoxia-, antioxidant- and apoptosis-related gene expression and apoptosis. Resveratrol significantly increased expression of VEGF, HIF-1α, Nrf2, CAT and Bcl-2 mRNA and decreased BAX and Caspase-3 mRNA and reduced numbers of TUNEL-positive cells after vitrification and heterotopic ovarian transplantation. Resveratrol improved the antioxidant capacity, reduced apoptosis and activated the HIF-1α/VEGF pathway to promote angiogenesis 3- and 9-days following transplantation. These results indicated that the addition of resveratrol to vitrification solutions intended for long-term cryopreservation of ovary tissues improves survival in storage and the grafts following transplantation. This study provides a theoretical basis for the successful transplantation of avian ovarian tissue after vitrification.
Collapse
Affiliation(s)
- Qingming Qin
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Zhili Li
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Rongxu Liu
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Shaoxia Liu
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Minghui Guo
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Min Zhang
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Haigang Wu
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China
| | - Li Huang
- Engineering and Technology Research Center for Waterfowl Resources Development and Utilization and Epidemic Disease Prevention and Control of Henan Province, Xinyang Agriculture and Forestry University, Xinyang, Henan Province, 464000, PR China.
| |
Collapse
|
3
|
Yong Q, Dijkstra KL, van der Keur C, Bruijn JA, Eikmans M, Baelde HJ. MIF Increases sFLT1 Expression in Early Uncomplicated Pregnancy and Preeclampsia. Int J Mol Sci 2023; 24:10050. [PMID: 37373198 DOI: 10.3390/ijms241210050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Insufficient immune tolerance during pregnancy is associated with pathological conditions such as preeclampsia (PE). Soluble fms-like tyrosine kinase-1 (sFLT1), which exerts a role in the late stage of PE, has shown its beneficial anti-inflammatory effects in inflammation-associated diseases. Macrophage migration inhibitory factor (MIF) was reported to upregulate sFLT1 production in experimental congenital diaphragmatic hernia. However, the placental sFLT1 expression in early uncomplicated pregnancy and whether MIF can regulate sFLT1 expression in uncomplicated and preeclamptic pregnancy are unclear. We collected first-trimester placentas and term placentas from uncomplicated and preeclamptic pregnancies to investigate sFLT1 and MIF expression in vivo. Primary cytotrophoblasts (CTBs) and a human trophoblast cell line (Bewo) were used to study the regulation of MIF on sFLT1 expression in vitro. In placentas from first-trimester pregnancy, we observed a high expression of sFLT1, specifically in extravillous trophoblasts (EVTs) and syncytiotrophoblast (STB) cells. MIF mRNA levels strongly correlated with sFLT1 expression in term placentas from preeclamptic pregnancies. In in vitro experiments, sFLT1 and MIF levels increased significantly in CTBs during their differentiation to EVTs and STBs, and MIF inhibitor (ISO-1) significantly reduced sFLT1 expression in a dose-dependent manner during this process. sFLT1 showed significant upregulation with increasing doses of MIF in Bewo cells. Our results show that sFLT1 is highly expressed at the maternal-fetal interface during early pregnancy and that MIF can increase sFLT1 expression in early uncomplicated pregnancy and PE, which suggests that sFLT1 plays an essential role in the modulation of inflammation in pregnancy.
Collapse
Affiliation(s)
- Qing Yong
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Kyra L Dijkstra
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Carin van der Keur
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jan A Bruijn
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
4
|
Niu M, Zhao F, Chen R, Li P, Bi L. The transient receptor potential channels in rheumatoid arthritis: Need to pay more attention. Front Immunol 2023; 14:1127277. [PMID: 36926330 PMCID: PMC10013686 DOI: 10.3389/fimmu.2023.1127277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/06/2023] [Indexed: 03/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is characterized by the augment of vascular permeability, increased inflammatory cells infiltration, dysregulated immune cells activation, pannus formation and unbearable pain hyperalgesia. Ca2+ affect almost every aspect of cellular functions, involving cell migration, signal transduction, proliferation, and apoptosis. Transient receptor potential channels (TRPs) as a type of non-selective permeable cation channels, can regulate Ca2+ entry and intracellular Ca2+ signal in cells including immune cells and neurons. Researches have demonstrated that TRPs in the mechanisms of inflammatory diseases have achieved rapid progress, while the roles of TRPs in RA pathogenesis and pain hyperalgesia are still not well understood. To solve this problem, this review presents the evidence of TRPs on vascular endothelial cells in joint swelling, neutrophils activation and their trans-endothelial migration, as well as their bridging role in the reactive oxygen species/TRPs/Ca2+/peptidyl arginine deiminases networks in accelerating citrullinated proteins formation. It also points out the distinct functions of TRPs subfamilies expressed in the nervous systems of joints in cold hyperalgesia and neuro-inflammation mutually influenced inflammatory pain in RA. Thus, more attention could be paid on the impact of TRPs in RA and TRPs are useful in researches on the molecular mechanisms of anti-inflammation and analgesic therapeutic strategies.
Collapse
Affiliation(s)
- Mengwen Niu
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rui Chen
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
5
|
The VEGF Inhibitor Soluble Fms-like Tyrosine Kinase 1 Does Not Promote AKI-to-CKD Transition. Int J Mol Sci 2022; 23:ijms23179660. [PMID: 36077058 PMCID: PMC9456014 DOI: 10.3390/ijms23179660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Soluble Fms-like tyrosine kinase 1 (sFLT1) is an endogenous VEGF inhibitor. sFLT1 has been described as an anti-inflammatory treatment for diabetic nephropathy and heart fibrosis. However, sFLT1 has also been related to peritubular capillary (PTC) loss, which promotes fibrogenesis. Here, we studied whether transfection with sFlt1 aggravates experimental AKI-to-CKD transition and whether sFLT1 is increased in human kidney fibrosis. (2) Methods: Mice were transfected via electroporation with sFlt1. After confirming transfection efficacy, mice underwent unilateral ischemia/reperfusion injury (IRI) and were sacrificed 28 days later. Kidney histology and RNA were analyzed to study renal fibrosis, PTC damage and inflammation. Renal sFLT1 mRNA expression was measured in CKD biopsies and control kidney tissue. (3) Results: sFlt1 transfection did not aggravate renal fibrosis, PTC loss or macrophage recruitment in IRI mice. In contrast, higher transfection efficiency was correlated with reduced expression of pro-fibrotic and pro-inflammatory markers. In the human samples, sFLT1 mRNA levels were similar in CKD and control kidneys and were not correlated with interstitial fibrosis or PTC loss. (4) Conclusion: As we previously found that sFLT1 has therapeutic potential in diabetic nephropathy, our findings indicate that sFLT1 can be administered at a dose that is therapeutically effective in reducing inflammation, without promoting maladaptive kidney damage.
Collapse
|
6
|
Hu Z, Xiao M, Cai H, Li W, Fang W, Long X. Glycyrrhizin regulates rat TMJOA progression by inhibiting the HMGB1-RAGE/TLR4-NF-κB/AKT pathway. J Cell Mol Med 2021; 26:925-936. [PMID: 34953035 PMCID: PMC8817133 DOI: 10.1111/jcmm.17149] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 12/06/2021] [Accepted: 12/12/2021] [Indexed: 11/26/2022] Open
Abstract
To investigate the role of glycyrrhizin on the progression of temporomandibular joint osteoarthritis (TMJOA) and the underlying mechanism by regulation of the high‐mobility group box 1 (HMGB1) receptor for advanced glycation end products (RAGE)/toll‐like receptor 4 (TLR4)‐nuclear factor kappa B (NF‐κB)/protein kinase B (AKT) pathway. After a rat model of TMJOA was built by intra‐articular injection of monosodium iodoacetate, glycyrrhizin was intragastrically administered at low concentration (20 mg/kg) or high concentration (50 mg/kg). Micro‐computed tomography, histological and immunohistochemical analysis were used to reveal the progression of TMJOA. Rat TMJ chondrocytes and disc cells were cultured in inflammatory condition with different doses of glycyrrhizin. Western blot was used to evaluate the effect of glycyrrhizin on the HMGB1‐RAGE/TLR4‐NF‐κB/AKT pathway. Administration of glycyrrhizin alleviated cartilage degeneration, lowered the levels of inflammatory and catabolic mediators and reduced the production of HMGB1, RAGE and TLR4 in TMJOA animal model. Increased production of RAGE and TLR4, and activated intracellular NF‐κB and/or AKT signalling pathways in chondrocytes and disc cells were found in inflammatory condition. Upon activation, matrix metalloprotease‐3 and interleukin‐6 were upregulated. Glycyrrhizin inhibited not only HMGB1 release but also RAGE and TLR4 in inflammatory condition. Glycyrrhizin alleviated the pathological changes of TMJOA by regulating the HMGB1‐RAGE/TLR4‐NF‐kB/AKT signalling pathway. This study revealed the potential of glycyrrhizin as a novel therapeutic drug to suppress TMJ cartilage degradation.
Collapse
Affiliation(s)
- Zhihui Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mian Xiao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Hengxing Cai
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Fang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Murdaca G, Allegra A, Paladin F, Calapai F, Musolino C, Gangemi S. Involvement of Alarmins in the Pathogenesis and Progression of Multiple Myeloma. Int J Mol Sci 2021; 22:9039. [PMID: 34445745 PMCID: PMC8396675 DOI: 10.3390/ijms22169039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE Multiple Myeloma (MM) is a haematological disease resulting from the neoplastic transformation of plasma cells. The uncontrolled growth of plasma cells in the bone marrow and the delivery of several cytokines causes bone erosion that often does not regress, even in the event of disease remission. MM is characterised by a multi-step evolutionary path, which starts with an early asymptomatic stage defined as monoclonal gammopathy of undetermined significance (MGUS) evolving to overt disease. DATA SOURCES AND STUDY SELECTION We have selected scientific publications on the specific topics "alarmis, MGUS, and MM", drawing from PubMed. The keywords we used were alarmines, MGUS, MM, and immune system. RESULTS The analysis confirms the pivotal role of molecules such as high-mobility group box-1, heat shock proteins, and S100 proteins in the induction of neoangiogenesis, which represents a milestone in the negative evolution of MM as well as other haematological and non-haematological tumours. CONCLUSIONS Modulation of the host immune system and the inhibition of neoangiogenesis may represent the therapeutic target for the treatment of MM that is capable of promoting better survival and reducing the risk of RRMM.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino IRCCS, 20132 Genoa, Italy;
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Francesca Paladin
- Department of Internal Medicine, University of Genoa, Ospedale Policlinico San Martino IRCCS, 20132 Genoa, Italy;
| | - Fabrizio Calapai
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98168 Messina, Italy;
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
8
|
Novel Blood Indicators of Progression and Prognosis in Renal Cell Carcinoma: Red Cell Distribution Width-to-Lymphocyte Ratio and Albumin-to-Fibrinogen Ratio. JOURNAL OF ONCOLOGY 2020; 2020:2895150. [PMID: 33299415 PMCID: PMC7710420 DOI: 10.1155/2020/2895150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Objective To evaluate the value of preoperative red cell distribution width-to-lymphocyte ratio (RLR) and albumin-to-fibrinogen ratio (AFR) to the prognosis of patients after renal cell carcinoma (RCC). Methods From 2012 to 2016, a total of 273 RCC patients underwent radical nephrectomy or partial nephrectomy. This study retrospectively analyzed this group of patients. X-tile software was used to determine the optimal values of RLR and AFR in the peripheral blood. The nomogram constructed with independent factors was used to predict the survival outcome of the patients after RCC. Results The RLR of the RCC group was higher than that of the normal control group (P=0.002), whereas the AFR of the RCC group was lower than that of the normal control group (P < 0.001). RLR and AFR are related to tumour type and tumour-node-metastasis (TNM) stage (P < 0.05 for all). Cox regression analysis showed that the independent prognostic factors affecting overall survival and disease-free survival in the RCC group were symptom, tumour type, TNM stage, Fuhrman grade, RLR, and AFR (P < 0.05 for all). The nomogram constructed by multiple factors has better predictive power for patients after RCC. Conclusion Preoperative RLR and AFR can serve as potential biomarkers to predict the prognosis of postoperative RCC patients and improve the predictability of patient recurrence and survival.
Collapse
|
9
|
Xiao Y, Ding L, Yin S, Huang Z, Zhang L, Mei W, Wu P, Wang P, Pan K. Relationship between the pyroptosis of fibroblast‑like synoviocytes and HMGB1 secretion in knee osteoarthritis. Mol Med Rep 2020; 23:97. [PMID: 33300062 PMCID: PMC7723153 DOI: 10.3892/mmr.2020.11736] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 09/11/2020] [Indexed: 11/23/2022] Open
Abstract
High mobility group box 1 (HMGB1) is an important downstream product of pyroptosis in macrophages, and it serves a vital role in numerous inflammatory diseases. Previous studies have reported that HMGB1 is released by fibroblast-like synoviocytes (FLSs) that are activated by inflammatory cytokines in knee osteoarthritis (KOA); however, the mechanism via which FLS promotes HMGB1 secretion in KOA remains unknown. According to our previous study, pyroptosis occurs in FLSs of patients with KOA and is mediated by Nod-like receptor protein (NLRP)1 or NLRP3 inflammasomes. However, the specific relationship between HMGB1 secretion and FLS pyroptosis requires further investigation. In the present study, the association between HMGB1 secretion and FLS pyroptosis was investigated in vitro and in vivo. In this study, western blotting, ELISA and reverse transcription-quantitative PCR were used to measure expression levels of proteins and mRNA. Caspase-1 activity assay and Hoechst 33342/PI double staining were used to observe the pyroptosis of FLSs. Hematoxylin and eosin staining was used to observe the destruction of cartilage in KOA. Increased expression levels of pyroptosis-related proteins and HMGB1 in the synovium of rat anterior cruciate ligament transection-induced KOA models were identified, and these changes were significantly mitigated via the intra-articular injection of a caspase-1 inhibitor. In vitro, FLSs were treated with lipopolysaccharide (LPS) + ATP to induce pyroptosis, and HMGB1 secretion was subsequently measured. LPS + ATP significantly increased the expression levels of pyroptosis-related proteins and HMGB1 in FLSs, and these effects were significantly mitigated by small interfering RNAs targeting NLRP1, NLRP3, apoptosis-associated speck-like protein with a caspase-recruitment domain or caspase-1. Therefore, the present results indicated that NLRP1/NLRP3 inflammasome-mediated and caspase-1-dependent FLS pyroptosis increased HMGB1 secretion in KOA. These findings may provide a therapeutic strategy to decrease synovial inflammatory responses during KOA progression.
Collapse
Affiliation(s)
- Yancheng Xiao
- Department of Orthopedics, Liyang Hospital of Traditional Chinese Medicine, Liyang, Jiangsu 213300, P.R. China
| | - Liang Ding
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Songjiang Yin
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Zhengquan Huang
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Li Zhang
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Wei Mei
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Peng Wu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Peimin Wang
- Department of Orthopedics, Jiangsu Province Hospital of Traditional Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Ke Pan
- Department of Orthopedics, Liyang Hospital of Traditional Chinese Medicine, Liyang, Jiangsu 213300, P.R. China
| |
Collapse
|
10
|
Gacaferi H, Mimpen JY, Baldwin MJ, Snelling SJB, Nelissen RGHH, Carr AJ, Dakin SG. The potential roles of high mobility group box 1 (HMGB1) in musculoskeletal disease: A systematic review. TRANSLATIONAL SPORTS MEDICINE 2020. [DOI: 10.1002/tsm2.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Hamez Gacaferi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
- Department of Orthopaedics Leiden University Medical Centre Leiden The Netherlands
| | - Jolet Y. Mimpen
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Mathew J. Baldwin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Sarah J. B. Snelling
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | | | - Andrew J. Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| | - Stephanie G. Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS) Botnar Research Centre University of Oxford Oxford UK
| |
Collapse
|
11
|
Zheng X, Lv Y, Li S, Zhang Q, Zhang X, Hao Z. Adeno-associated virus-mediated colonic secretory expression of HMGB1 A box attenuates experimental colitis in mice. J Gene Med 2018; 18:261-272. [PMID: 27572454 DOI: 10.1002/jgm.2899] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Extracellular high mobility group box 1 (HMGB1) is crucially implicated in the pathogenesis of inflammatory bowel diseases (IBDs). A box domain of HMGB1 has been identified as a specific antagonist of HMGB1. In the present study, we tested the effects of adeno-associated virus (AAV)-mediated colonic secretory expression of HMGB1 A box on murine experimental colitis. METHODS Self-complementary AAV-2 carrying mouse immunoglobin Gκ leader-human HMGB1 A box (AAV-HMGB1 A box) was constructed. The effects of intracolonically administered AAV-HMGB1 A box on dextran sulfate sodium (DSS)- and 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis were assessed by the disease activity index (DAI), colon length, macroscopic and histological scoring, myeloperoxidase (MPO) activity, and epithelial apoptosis and complementary proliferation. Colonic immune cell infiltrates, mucosal malondialdehyde content and superoxide dismutase activity, colonic tumor necrosis factor-α (TNF-α), interleukin (IL)-1β and IL-10 levels, serum HMGB1 concentration, and colonic HMGB1 release were determined to investigate the underlying mechanisms. RESULTS Intracolonically administered AAV-HMGB1 A box efficiently mediated secretory expression of HMGB1 A box and led to significant decreases in DAI, macroscopic and histological scores and colonic epithelial apoptosis in both DSS- and TNBS-treated mice. Modulating inflammation-associated cytokines, such as inhibiting colonic TNF-α and IL-1β expression, decreasing HMGB1 release, and restoring colonic IL-10 levels, and thereby inhibiting inflammatory cell infiltration and alleviating oxidant damage, might be the underlying mechanism. CONCLUSIONS Intracolonic application of AAV-HMGB1 A box is effective in alleviating murine colitis and has therapeutic potential in human IBDs.
Collapse
Affiliation(s)
- Xiaoyan Zheng
- Department of Rheumatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yifei Lv
- Department of Gastroenterology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi Province, People's Republic of China
| | - Shuang Li
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Qiannan Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xueting Zhang
- Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Zhiming Hao
- Department of Rheumatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China. .,Department of Gastroenterology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China.
| |
Collapse
|
12
|
Feng Y, Ke J, Cao P, Deng M, Li J, Cai H, Meng Q, Li Y, Long X. HMGB1-induced angiogenesis in perforated disc cells of human temporomandibular joint. J Cell Mol Med 2017; 22:1283-1291. [PMID: 29083089 PMCID: PMC5783830 DOI: 10.1111/jcmm.13410] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/29/2017] [Indexed: 11/29/2022] Open
Abstract
High mobility group 1 protein (HMGB1), a highly conserved nuclear DNA‐binding protein and inflammatory mediator, has been recently found to be involved in angiogenesis. Our previous study has demonstrated the elevation of HMGB1 in the tissue of perforated disc of temporomandibular joint (TMJ). Here, we investigated a novel mediator of HMGB1 in regulating hypoxia‐inducible factor‐1α (HIF‐1α) and vascular endothelial growth factor (VEGF) to mediate angiogenesis in perforated disc cells of TMJ. HMGB1 increased the expression of HIF‐1α and VEGF in a dose‐ and time‐dependent manner in these cells. Moreover, immunofluorescence assay exhibits that the HIF‐1α were activated by HMGB1. In addition, HMGB1 activated extracellular signal‐related kinase 1/2 (Erk1/2), Jun N‐terminal kinase (JNK), but not P38 in these cells. Furthermore, both U0126 (ErK inhibitor) and SP600125 (JNK inhibitor) significantly suppressed the enhanced production of HIF‐1α and VEGF induced by HMGB1. Tube formation of human umbilical vein endothelial cells (HUVECs) was significantly increased by exposure to conditioned medium derived from HMGB1‐stimulated perforated disc cells, while attenuated with pre‐treatment of inhibitors for VEGF, HIF‐1α, Erk and JNK, individually. Therefore, abundance of HMGB1 mediates activation of HIF‐1α in disc cells via Erk and JNK pathway and then, initiates VEGF secretion, thereby leading to disc angiogenesis and accelerating degenerative change of the perforated disc.
Collapse
Affiliation(s)
- Yaping Feng
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jin Ke
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Pinyin Cao
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST), Key Laboratory of Oral Biomedicine Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Mohong Deng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Jian Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Hengxing Cai
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Qinggong Meng
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Yingjie Li
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Xing Long
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
13
|
The Role of High-Mobility Group Box-1 and Its Crosstalk with Microbiome in Rheumatoid Arthritis. Mediators Inflamm 2017; 2017:5230374. [PMID: 29200665 PMCID: PMC5672636 DOI: 10.1155/2017/5230374] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 07/16/2017] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic, definitely disabling, and potentially severe autoimmune disease. Although an increasing number of patients are affected, a key treatment for all patients has not been discovered. High-mobility group box-1 (HMGB1) is a nuclear protein passively and actively released by almost all cell types after several stimuli. HMGB1 is involved in RA pathogenesis, but a convincing explanation about its role and possible modulation in RA is still lacking. Microbiome and its homeostasis are altered in patients with RA, and the microbiota restoration has been proposed to patients with RA. The purpose of the present review is to analyze the available evidences regarding HMGB1 and microbiome roles in RA and the possible implications of the crosstalk between the nuclear protein and microbiome in understanding and possibly treating patients affected by this harmful condition.
Collapse
|
14
|
Biscetti F, Gentileschi S, Bertucci F, Servillo M, Arena V, Angelini F, Stigliano E, Bonanno G, Scambia G, Sacchetti B, Pierelli L, Landolfi R, Flex A. The angiogenic properties of human adipose-derived stem cells (HASCs) are modulated by the High mobility group box protein 1 (HMGB1). Int J Cardiol 2017; 249:349-356. [PMID: 28967436 DOI: 10.1016/j.ijcard.2017.09.165] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 09/04/2017] [Accepted: 09/18/2017] [Indexed: 12/16/2022]
Abstract
Peripheral arterial disease (PAD), is a major health problem. Many studies have been focused on the possibilities of treatment offered by vascular regeneration. Human adipose-derived stem cells (HASCs), multipotent CD34+ stem cells found in the stromal-vascular fraction of adipose tissues, which are capable to differentiate into multiple mesenchymal cell types. The High mobility group box 1 protein (HMGB1) is a nuclear protein involved in angiogenesis. The aim of the study was to define the role of HMGB1 in cell therapy with HASCs, in an animal model of PAD. We induced unilateral ischemia in mice and we treated them with HASCs, with the specific HMGB1-inihibitor BoxA, with HMGB1 protein, and with the specific VEGF inhibitor sFlt1, alternately or concurrently. We measured the blood flow recovery in all mice. Immunohistochemical and ELISA analyses was performed to evaluate the number of vessels and the VEGF tissue content. None auto-amputation occurred and there have been no rejection reactions to the administration of HASCs. Animals co-treated with HASCs and HMGB1 protein had an improved blood flow recovery, compared to HASCs-treated mice. The post-ischemic angiogenesis was reduced when the HMGB1 pathway was blocked or when the VEGF activity was inhibited, in mice co-treated with HASCs and HMGB1. In conclusion, the HASCs treatment can be used in a mouse model of PAD to induce post-ischemic angiogenesis, modulating angiogenesis by HMGB1. This effect is mediated by VEGF activity. Although further data are needed, these findings shed light on possible new cell treatments for patients with PAD.
Collapse
Affiliation(s)
- Federico Biscetti
- Division of Rheumatology, Institute of Rheumatology & Related Sciences, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy; Laboratory of Vascular Biology and Genetics, Department of Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy.
| | - Stefano Gentileschi
- Division of Plastic Surgery, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Flavio Bertucci
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Maria Servillo
- Division of Plastic Surgery, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Vincenzo Arena
- Department of Pathology, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Flavia Angelini
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Egidio Stigliano
- Department of Pathology, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Giuseppina Bonanno
- Division of Gynecology, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Giovanni Scambia
- Division of Gynecology, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | | | - Luca Pierelli
- Immunohematology and Transfusion Medicine, San Camillo Forlanini Hospital, Rome, Italy; Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Raffaele Landolfi
- Department of Internal Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| | - Andrea Flex
- Laboratory of Vascular Biology and Genetics, Department of Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy; Department of Internal Medicine, Fondazione Policlinico Universitario "A. Gemelli", Catholic University School of Medicine, Rome, Italy
| |
Collapse
|
15
|
Bus P, Scharpfenecker M, Van Der Wilk P, Wolterbeek R, Bruijn JA, Baelde HJ. The VEGF-A inhibitor sFLT-1 improves renal function by reducing endothelial activation and inflammation in a mouse model of type 1 diabetes. Diabetologia 2017; 60. [PMID: 28620823 PMCID: PMC5552850 DOI: 10.1007/s00125-017-4322-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS Animal models of diabetic nephropathy show increased levels of glomerular vascular endothelial growth factor (VEGF)-A, and several studies have shown that inhibiting VEGF-A in animal models of diabetes can prevent albuminuria and glomerular hypertrophy. However, in those studies, treatment was initiated before the onset of kidney damage. Therefore, the aim of this study was to investigate whether transfecting mice with the VEGF-A inhibitor sFlt-1 (encoding soluble fms-related tyrosine kinase 1) can reverse pre-existing kidney damage in a mouse model of type 1 diabetes. In addition, we investigated whether transfection with sFlt-1 can reduce endothelial activation and inflammation in these mice. METHODS Subgroups of untreated 8-week-old female C57BL/6J control (n = 5) and diabetic mice (n = 7) were euthanised 5 weeks after the start of the experiment in order to determine the degree of kidney damage prior to treatment with sFLT-1. Diabetes was induced with three i.p. injections of streptozotocin (75 mg/kg) administered at 2 day intervals. Diabetic nephropathy was then investigated in diabetic mice transfected with sFlt-1 (n = 6); non-diabetic, non-transfected control mice (n = 5); non-diabetic control mice transfected with sFlt-1(n = 10); and non-transfected diabetic mice (n = 6). These mice were euthanised at the end of week 15. Transfection with sFlt-1 was performed in week 6. RESULTS We found that transfection with sFlt-1 significantly reduced kidney damage by normalising albuminuria, glomerular hypertrophy and mesangial matrix content (i.e. glomerular collagen type IV protein levels) (p < 0.001). We also found that transfection with sFlt-1 reduced endothelial activation (p < 0.001), glomerular macrophage infiltration (p < 0.001) and glomerular TNF-α protein levels (p < 0.001). Finally, sFLT-1 decreased VEGF-A-induced endothelial activation in vitro (p < 0.001). CONCLUSIONS/INTERPRETATION These results suggest that sFLT-1 might be beneficial in treating diabetic nephropathy by inhibiting VEGF-A, thereby reducing endothelial activation and glomerular inflammation, and ultimately reversing kidney damage.
Collapse
Affiliation(s)
- Pascal Bus
- Department of Pathology, Leiden University Medical Center, L1Q, Room P0-107, P.O. Box 9600, 2300 RC, Leiden, the Netherlands.
| | - Marion Scharpfenecker
- Department of Pathology, Leiden University Medical Center, L1Q, Room P0-107, P.O. Box 9600, 2300 RC, Leiden, the Netherlands
| | - Priscilla Van Der Wilk
- Department of Pathology, Leiden University Medical Center, L1Q, Room P0-107, P.O. Box 9600, 2300 RC, Leiden, the Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan A Bruijn
- Department of Pathology, Leiden University Medical Center, L1Q, Room P0-107, P.O. Box 9600, 2300 RC, Leiden, the Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, L1Q, Room P0-107, P.O. Box 9600, 2300 RC, Leiden, the Netherlands
| |
Collapse
|
16
|
Jiang Z, Zhou Q, Gu C, Li D, Zhu L. Depletion of circulating monocytes suppresses IL-17 and HMGB1 expression in mice with LPS-induced acute lung injury. Am J Physiol Lung Cell Mol Physiol 2016; 312:L231-L242. [PMID: 27913426 DOI: 10.1152/ajplung.00389.2016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 12/26/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is an important cause of mortality in critically ill patients. Macrophages play an important role in the pathogenesis of ALI/ARDS. To investigate the role and underlying mechanisms of circulating monocytes and resident alveolar macrophages (AMs) in ALI/ARDS, we depleted circulating monocytes and AMs by clodronate-loaded liposome (CL) in a lipopolysaccharide (LPS)-induced ALI/ARDS mouse model. Our results indicated that depletion of circulating monocytes by intravenous injection of CL 2 days before intratracheal LPS treatment significantly suppressed the acute lung injury in mice with ALI/ARDS, accompanied with significant reduction in neutrophil influx, interleukin-17, monocyte chemoattractant protein 1, high-mobility group box 1 protein, suppressor of cytokine signaling 3, and surfactant protein D (SP-D) in the lungs of 2 days intratracheal LPS-treated mice. In contrast, depletion of AMs by intratracheal delivery of CL enhanced the acute lung injury in association with upregulation of these mediators. Blocking monocyte chemoattractant protein 1 signaling by intraperitoneal instillation of anti-mouse CCL2 neutralizing antibody significantly reduced acute lung injury and neutrophil influx. In addition, SP-D was upregulated by mediators released from AMs because primary murine type II alveolar epithelial cells expressed more SP-D after treatment with bronchoalveolar lavage from LPS-treated mice or the conditioned media from LPS-treated RAW 264.7 cells. The results indicated that circulating monocytes are proinflammatory, but AMs have anti-inflammatory functions in the early phase of ALI/ARDS. The study provided a molecular basis for the treatment of ALI/ARDS through modulation of circulating monocytes and AMs.
Collapse
Affiliation(s)
- Zhilong Jiang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qianlin Zhou
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chenlin Gu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dandan Li
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Zhu
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Zou L, Zhang G, Liu L, Chen C, Cao X, Cai J. Relationship between PI3K pathway and angiogenesis in CIA rat synovium. Am J Transl Res 2016; 8:3141-3147. [PMID: 27508035 PMCID: PMC4969451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/30/2016] [Indexed: 06/06/2023]
Abstract
To investigate the expression of hypoxia inducible factor (HIF-1α) and vascular endothelial growth factor (VEGF) in the synovium of collagen-induced arthritis (CIA) joint, and whether the PI3K pathway regulates angiogenesis in rheumatoid arthritis or not. A randomized controlled according to the principle of the rats were divided into normal control group (10 rats) and the experimental group (40 rats). The experimental group rats were established as type II collagen plus adjuvant Freund's complete adjuvant-induced arthritis model. HIF-1α and VEGF proteins' expression in serum of CIA rats group and normal control group were detected by ELISA. Microvessel density (MVD) in synovial tissue of CIA rats group and normal control group were detected by immunohistochemistry (IHC) staining. The protein expression of PTEN, PI3K, and AKT in synovial tissue were detected by Western Blot. Compared with normal control group, toes and ankle swelling and arthritis index (AI) of CIA rat increased, and the expression of VEGF and HIF-1α proteins in peripheral serum increased, IHC showed that MVD was significantly higher than that of the control group, and the difference was statistically significant (p<0.05). Western Blot results showed that PI3K and AKT proteins expression in CIA synovial tissue of rats increased, while the expression of PTEN protein decreased. Correlation analysis showed that VEGF and HIF-1 levels in the peripheral serum of CIA rats were positively correlated with arthritis index (AI); the contents of HIF-1α and VEGF in the peripheral serum of CIA rats were positively correlated with MVD in synovium tissue. The CIA rat model regulated the expression of HIF-1α and VEGF proteins in peripheral serum by PI3K signaling pathway, and then regulated neovascularization in RA.
Collapse
Affiliation(s)
- Lin Zou
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| | - Guichun Zhang
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| | - Lifeng Liu
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| | - Chen Chen
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| | - Xuecheng Cao
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| | - Jinfang Cai
- Department of Traumatic Orthopedic Surgery, The General Hospital of Jinan Military Command Jinan 250031, Shandong, China
| |
Collapse
|