1
|
Prantner D, Vogel SN. Intracellular methylglyoxal accumulation in classically activated mouse macrophages is mediated by HIF-1α. J Leukoc Biol 2025; 117:qiae215. [PMID: 39360990 DOI: 10.1093/jleuko/qiae215] [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: 08/08/2024] [Accepted: 09/30/2024] [Indexed: 03/30/2025] Open
Abstract
Approximately one million cases of sepsis in the United States occur annually. The early phase of sepsis features dramatic changes in host metabolism and inflammation. While examining the effects of metabolic pathways on inflammation, we discovered that the highly reactive glycolytic metabolite, methylglyoxal (MG), accumulates intracellularly during classical activation of macrophages. Herein, we explored the role of glycolysis and the master regulator of glycolysis, Hypoxia-Inducing Factor-1α (HIF-1α), in inflammation and MG accumulation in mouse and human macrophages. To determine how HIF-1α regulates the inflammatory response of macrophages, we correlated HIF-1α stabilization with proinflammatory gene expression and MG-adduct accumulation in WT vs HIF1a-deficient macrophages treated with LPS or LPS + IFN-γ. A nearly complete loss of HIF-1α protein expression in response to the hypoxia mimetic, cobalt chloride, confirmed the phenotype of the HIF1a-deficient macrophages. Moreover, absence of HIF-1α was also associated with decreased MG accumulation. Increasing the glucose concentration in cultured macrophages was sufficient to cause accumulation of endogenous MG-adducts which correlated with increased Tnf and Il1b expression during classical activation. The use of the MG antagonist, aminoguanidine, led to a significant decrease in Tnf and Il1b expression in both mouse macrophages and the THP-1 human macrophage cell line. Although off-target effects cannot be ruled out, these results are consistent with the possibility that MG regulates cytokine expression in classically activated macrophages. Collectively, this work suggests that HIF-1α stabilization is upstream of MG accumulation and that targeting the activity of HIF-1α in macrophages may be therapeutic during sepsis by limiting endogenous MG accumulation.
Collapse
Affiliation(s)
- Daniel Prantner
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore St., Suite 380, Baltimore, MD 21201, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore St., Suite 380, Baltimore, MD 21201, USA
| |
Collapse
|
2
|
Song J, Han S, Amaru R, Lanikova L, Quispe T, Kim D, Crawford JE, Kim SJ, Lee Y, Prchal JT. Alternatively spliced NFKB1 transcripts enriched in Andean Aymara modulate inflammation, HIF and hemoglobin. Nat Commun 2025; 16:1766. [PMID: 39971917 PMCID: PMC11840074 DOI: 10.1038/s41467-025-56848-0] [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: 09/22/2023] [Accepted: 02/03/2025] [Indexed: 02/21/2025] Open
Abstract
The molecular basis of increased hemoglobin in Andean Aymara highlanders is unknown. We conducted an integrative analysis of whole-genome-sequencing and granulocytes transcriptomics from Aymara and Europeans in Bolivia to explore genetic basis of the Aymara high hemoglobin. Differentially expressed and spliced genes in Aymaras were associated with inflammatory and hypoxia-related pathways. We identified transcripts with 4th or 5th exon skipping of NFKB1 (AS-NFKB1), key part of NF-kB complex, and their splicing quantitative trait loci; these were increased in Aymaras. AS-NFKB1 transcripts correlated with both transcripts and protein levels of inflammatory and HIF-regulated genes, including hemoglobin. While overexpression of the AS-NFKB1 variant led to increased expression of inflammatory and HIF-targeted genes; under inflammatory stress, NF-kB protein translocation to the nucleus was attenuated, resulting in reduced expression of these genes. Our study reveals AS-NFKB1 splicing events correlating with increased hemoglobin in Aymara and their possible protective mechanisms against excessive inflammation.
Collapse
Affiliation(s)
- Jihyun Song
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah and VA Hospital, Salt Lake City, UT, USA
| | - Seonggyun Han
- Department of Biomedical Informatics, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Psychiatry, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | - Ricardo Amaru
- Cell Biology Unit, School of Medicine, San Andres University, National Academy of Sciences, La Paz, Bolivia
| | - Lucie Lanikova
- Department of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Teddy Quispe
- Cell Biology Unit, School of Medicine, San Andres University, National Academy of Sciences, La Paz, Bolivia
| | - Dongwook Kim
- Department of Biomedical Informatics, School of Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Soo Jin Kim
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah and VA Hospital, Salt Lake City, UT, USA
| | - Younghee Lee
- College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul, Republic of Korea.
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, Huntsman Cancer Institute, University of Utah and VA Hospital, Salt Lake City, UT, USA.
| |
Collapse
|
3
|
Ruggieri E, Di Domenico E, Locatelli AG, Isopo F, Damanti S, De Lorenzo R, Milan E, Musco G, Rovere-Querini P, Cenci S, Vénéreau E. HMGB1, an evolving pleiotropic protein critical for cellular and tissue homeostasis: Role in aging and age-related diseases. Ageing Res Rev 2024; 102:102550. [PMID: 39427887 DOI: 10.1016/j.arr.2024.102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Aging is a universal biological process characterized by a progressive, cumulative decline in homeostatic capabilities and physiological functions, which inevitably increases vulnerability to diseases. A number of molecular pathomechanisms and hallmarks of aging have been recognized, yet we miss a thorough understanding of their complex interconnectedness. This review explores the molecular and cellular mechanisms underlying human aging, with a focus on the multiple roles of high mobility group Box 1 protein (HMGB1), the archetypal damage-associated molecular pattern (DAMP) molecule. In the nucleus, this non-histone chromatin-associated protein functions as a DNA chaperone and regulator of gene transcription, influencing DNA structure and gene expression. Moreover, this versatile protein can translocate to the cytoplasm to orchestrate other processes, such as autophagy, or be unconventionally secreted into the extracellular environment, where it acts as a DAMP, combining inflammatory and regenerative properties. Notably, lower expression of HMGB1 within the cell and its heightened extracellular release have been associated with diverse age-associated traits, making it a suitable candidate as a universal biomarker of aging. In this review, we outline the evidence implicating HMGB1 in aging, also in light of an evolutionary perspective on its functional pleiotropy, and propose critical issues that need to be addressed to gauge the value of HMGB1 as a potential biomarker across age-related diseases and therapeutic target to promote healthy longevity.
Collapse
Affiliation(s)
- Elena Ruggieri
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Erika Di Domenico
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Flavio Isopo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Enrico Milan
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| | - Emilie Vénéreau
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
4
|
Zhao L, Wu Q, Long Y, Qu Q, Qi F, Liu L, Zhang L, Ai K. microRNAs: critical targets for treating rheumatoid arthritis angiogenesis. J Drug Target 2024; 32:1-20. [PMID: 37982157 DOI: 10.1080/1061186x.2023.2284097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/09/2023] [Indexed: 11/21/2023]
Abstract
Vascular neogenesis, an early event in the development of rheumatoid arthritis (RA) inflammation, is critical for the formation of synovial vascular networks and plays a key role in the progression and persistence of chronic RA inflammation. microRNAs (miRNAs), a class of single-stranded, non-coding RNAs with approximately 21-23 nucleotides in length, regulate gene expression by binding to the 3' untranslated region (3'-UTR) of specific mRNAs. Increasing evidence suggests that miRNAs are differently expressed in diseases associated with vascular neogenesis and play a crucial role in disease-related vascular neogenesis. However, current studies are not sufficient and further experimental studies are needed to validate and establish the relationship between miRNAs and diseases associated with vascular neogenesis, and to determine the specific role of miRNAs in vascular development pathways. To better treat vascular neogenesis in diseases such as RA, we need additional studies on the role of miRNAs and their target genes in vascular development, and to provide more strategic references. In addition, future studies can use modern biotechnological methods such as proteomics and transcriptomics to investigate the expression and regulatory mechanisms of miRNAs, providing a more comprehensive and in-depth research basis for the treatment of related diseases such as RA.
Collapse
Affiliation(s)
- Lingyun Zhao
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Qingze Wu
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Yiying Long
- Hunan Traditional Chinese Medical College, Zhuzhou, China
| | - Qirui Qu
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Fang Qi
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Li Liu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Liang Zhang
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| | - Kun Ai
- College of Acupuncture, Tuina and Rehabilitation, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
5
|
Zhao Y, Li H, Ma X, Meng X, Tang Q. Identification of biomarkers related to angiogenesis in myocardial ischemia-reperfusion injury and prediction of potential drugs. PLoS One 2024; 19:e0300790. [PMID: 38935597 PMCID: PMC11210787 DOI: 10.1371/journal.pone.0300790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 03/05/2024] [Indexed: 06/29/2024] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) refers to the secondary damage to myocardial tissue that occurs when blood perfusion is rapidly restored following myocardial ischemia. This process often exacerbates the injury to myocardial fiber structure and function. The activation mechanism of angiogenesis is closely related to MIRI and plays a significant role in the occurrence and progression of ischemic injury. In this study, we utilized sequencing data from the GEO database and employed WGCNA, Mfuzz cluster analysis, and protein interaction network to identify Stat3, Rela, and Ubb as hub genes involved in MIRI-angiogenesis. Additionally, the GO and KEGG analysis of differentially expressed genes highlighted their broad participation in inflammatory responses and associated signaling pathways. Moreover, the analysis of sequencing data and hub genes revealed a notable increase in the infiltration ratio of monocytes and activated mast cells. By establishing key cell ROC curves, using independent datasets, and validating the expression of hub genes, we demonstrated their high diagnostic value. Moreover, by scrutinizing single-cell sequencing data alongside trajectory analysis, it has come to light that Stat3 and Rela exhibit predominant expression within Dendritic cells. In contrast, Ubb demonstrates expression across multiple cell types, with all three genes being expressed at distinct stages of cellular development. Lastly, leveraging the CMap database, we predicted potential small molecule compounds for the identified hub genes and validated their binding activity through molecular docking. Ultimately, our research provides valuable evidence and references for the early diagnosis and treatment of MIRI from the perspective of angiogenesis.
Collapse
Affiliation(s)
- Yaowei Zhao
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Hongyu Li
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiyuan Ma
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xianghong Meng
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Qiang Tang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
6
|
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
|
7
|
Oda H, Nagamatsu T, Iriyama T, Osuga Y. Altered release of thrombomodulin and HMGB1 in the placenta complicated with preeclampsia. Placenta 2024; 148:12-19. [PMID: 38330539 DOI: 10.1016/j.placenta.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 10/31/2023] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
INTRODUCTION Preeclampsia (PE) is a severe pregnancy complication due to placental dysfunction. Thrombomodulin (TM), a glycoprotein expressed on the trophoblast cell membrane, plays an organ-protective role in the placenta by regulating coagulation and inflammation. TM-mediated regulation of High Mobility Group Box1(HMGB1) is an essential mechanism that contributes to placental homeostasis and prevents pregnancy complications in mice. Here, we aimed to clarify the role of placental TM and HMGB1 in the pathophysiology of human PE. METHODS AND RESULTS In this study, maternal blood serum and placental tissue were obtained from 72 PE patients and 110 normal controls. Soluble TM(sTM) and HMGB1 levels in the maternal serum were assessed. The placental TM and HMGB1 expression levels were evaluated using immunohistochemistry and qPCR. Serum sTM and HMGB1 levels gradually increased with gestational age in normal pregnancies; however, both circulating sTM and HMGB1 levels were significantly higher in the PE group. Serum HMGB1/sTM ratio was elevated in PE patients compared to that in normal controls, which correlated positively with the clinical severity of PE. The immunohistochemistry analysis revealed the loss of TM and the increase in extranuclear HMGB1. TM mRNA expression was diminished in PE placentas, which negatively correlated with soluble fms-like tyrosine kinase-1 (sFlt-1) expression. DISCUSSION The increase in circulating sTM and HMGB1 could be attributed to the enhanced placental TM shedding in PE patients. The molecular events mediated by the imbalance in the placental TM and HMGB1 levels could be an underlying feature of PE; maternal serum HMGB1/sTM ratio could reflect this status.
Collapse
Affiliation(s)
- Hiroko Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan; Department of Obstetrics and Gynecology, International University of Health and Welfare, Chiba, Japan.
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
8
|
Wei Z, Li H, Lv S, Yang J. Current situation and trend of non-coding RNA in rheumatoid arthritis: a review and bibliometric analysis. Front Immunol 2024; 14:1301545. [PMID: 38292492 PMCID: PMC10824985 DOI: 10.3389/fimmu.2023.1301545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 12/28/2023] [Indexed: 02/01/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic, systemic autoimmune disease that affects multiple joints and has adverse effects on various organs throughout the body, often leading to a poor prognosis. Recent studies have shown significant progress in the research of non-coding RNAs (ncRNAs) in RA. Therefore, this study aims to comprehensively assess the current status and research trends of ncRNAs in RA through a bibliometric analysis. Methods This study retrieved articles relevant to ncRNAs and RA from the Science Citation Index Expanded Database of the Web of Science Core Collection between January 1st, 2003, and July 31st, 2023. The relevant articles were screened based on the inclusion criteria. VOSviewer and CiteSpace are utilized for bibliometric and visual analysis. Results A total of 1697 publications were included in this study, and there was a noticeable increase in annual publications from January 1st, 2003, to July 31st, 2023. China, the United States, and the United Kingdom were the most productive countries in this field, contributing to 43.81%, 13.09%, and 3.87% of the publications. Anhui Medical University and Lu Qianjin were identified as the most influential institution and author. Frontiers In Immunology stood out as the most prolific journal, while Arthritis & Rheumatology was the most co-cited journal. Additionally, the research related to "circular RNA", "oxidative stress", "proliferation", and "migration" have emerged as new hotspots in the field. Conclusion In this study, we have summarized the publication characteristics related to ncRNA and RA and identified the most productive countries, institutions, authors, journals, hot topics, and trends.
Collapse
Affiliation(s)
- Zehong Wei
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Huaiyu Li
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Senhao Lv
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Junping Yang
- Clinical Laboratory, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Xu Q, Kong H, Ren S, Meng F, Liu R, Jin H, Zhang J. Coix seed oil alleviates synovial angiogenesis through suppressing HIF-1α/VEGF-A signaling pathways via SIRT1 in collagen-induced arthritis rats. Chin Med 2023; 18:119. [PMID: 37715217 PMCID: PMC10504826 DOI: 10.1186/s13020-023-00833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by symmetric arthritis. Coix Seed Oil (CSO) has been shown to reduce inflammation in collagen induced arthritis (CIA) rats. However, the effect of CSO on synovial angiogenesis in RA is unknown. In this study, we aimed to explore whether CSO could inhibit RA synovial angiogenesis and elucidate the underlying mechanisms. METHODS CIA rat models were established and subjected to different doses of CSO treatments for four weeks in vivo. Arthritis index, paw swelling, and weight were recorded to assess clinical symptoms. Hematoxylin and Eosin staining, Safarnin O fast green staining, Micro-CT, Immunohistochemical, and Immunofluorescence (IF) staining were performed to examined changes in synovial and joint tissues. The serum HIF-1α and VEGF-A levels were evaluated through enzyme-linked immunosorbent assay. Fibroblast-like synoviocytes (FLS) of rats was stimulated with tumor necrosis factor-α (TNF-α) for developing inflammatory model in vitro. Optimal concentrations of CSO and TNF-α for stimulation were measured through Cell Counting Kit-8 test. Wound healing and Transwell migration experiments were employed to determine FLS migratory ability. IF staining was performed to assess HIF-1α nuclear translocation in FLS. Protein levels of SIRT1, HIF-1α, VEGF-A, and CD31 were assessed through Western blot. The isolated aortic rings were induced with recombinant rat VEGF-A 165 (VEGF-A165) to observe the CSO inhibitory impact on angiogenesis ex vivo. RESULTS CSO attenuated the progression of arthritis in CIA rats, mitigated histopathological deterioration in synovial and joint tissues, significantly inhibited immature vessels labeled with CD31+/αSMA-, and reduced the micro-vessels in VEGF-A165 induced aortic rings. Moreover, it upregulated SIRT1 protein levels in CIA rats and TNF-α induced FLS, but decreased HIF-1α and VEGF-A protein levels. Furthermore, CSO inhibited the migration ability and HIF-1α nuclear translocation of TNF-α induced FLS. Finally, suppressing SIRT1 levels in TNF-α induced FLS enhanced their migration ability, HIF-1α nuclear translocation, and the protein levels of HIF-1α, VEGF-A, and CD31, whereas the inhibitory effect of CSO on TNF-α induced FLS was severely constrained. CONCLUSIONS This study indicates that CSO can alleviate synovial angiogenesis through suppressing HIF-1α/VEGF-A signaling pathways via SIRT1 in CIA rats.
Collapse
Affiliation(s)
- Qiangqiang Xu
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Hongxi Kong
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Shuang Ren
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Fanyan Meng
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Ruoshi Liu
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China
| | - Hongxin Jin
- Guangzhou University of Traditional Chinese Medicine, Guangdong, 510006, China
| | - Jie Zhang
- Department of Chinese Medicine, The First Hospital of China Medical University, Liaoning, 110001, China.
| |
Collapse
|
10
|
Ni L, Wang X, Xu G. Photoacoustic clinical applications: Musculoskeletal and abdominal imaging. Z Med Phys 2023; 33:324-335. [PMID: 37365088 PMCID: PMC10517401 DOI: 10.1016/j.zemedi.2023.04.009] [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: 11/22/2022] [Revised: 04/04/2023] [Accepted: 04/21/2023] [Indexed: 06/28/2023]
Abstract
Photoacoustic (PA) imaging has been extensively investigated in application in biomedicine over the last decade. This article reviews the motivation, significance, and system configuration of a few ongoing studies of implementing photoacoustic technology in musculoskeletal imaging, abdominal imaging, and interstitial sensing. The review then summarizes the methodologies and latest progress of relevant projects. Finally, we discuss our expectations for the future of translation research in PA imaging.
Collapse
Affiliation(s)
- Linyu Ni
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA
| | - Xueding Wang
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA
| | - Guan Xu
- Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, USA; Department of Ophthalmology and Visual Sciences, University of Michigan, 1000 Wall St., Ann Arbor, MI 48105, USA.
| |
Collapse
|
11
|
Chang Q, Fujio M, Tsuboi M, Bian H, Wakasugi M, Hibi H. High-mobility group box 1 accelerates distraction osteogenesis healing via the recruitment of endogenous stem/progenitor cells. Cytotherapy 2023:S1465-3249(23)00960-X. [PMID: 37354151 DOI: 10.1016/j.jcyt.2023.05.013] [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: 11/10/2022] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AIMS While distraction osteogenesis (DO) achieves substantial bone regeneration, prolonged fixation may lead to infections. Existing stem cell and physical therapies have limitations, requiring the development of novel therapeutic approaches. Here, we evaluated high-mobility group box 1 (HMGB1) as a novel therapeutic target for DO treatment. METHODS Micro-computed tomography (Micro-CT) analysis and histological staining of samples obtained from tibial DO model mice was performed. Transwell migration, wound healing, and proliferation assays were also performed on cultured human mesenchymal stem cells (hMSCs) and human umbilival vein endothelial cells (HUVECs). Tube formation assay was performed on HUVECs, whereas osteogenic differentiation assay was performed on hMSCs. RESULTS Micro-CT analysis and histological staining of mouse samples revealed that HMGB1 promotes bone regeneration during DO via the recruitment of PDGFRα and Sca-1 positve (PαS+) cells and endothelial progenitor cells. Furthermore, HMGB1 accelerated angiogenesis during DO, promoted the migration and osteogenic differentiation of hMSCs as well as the proliferation, migration and angiogenesis of HUVECs in vitro. CONCLUSIONS Our findings suggest that HMGB1 has a positive influence on endogenous stem/progenitor cells, representing a novel therapeutic target for the acceleration of DO-driven bone regeneration.
Collapse
Affiliation(s)
- Qi Chang
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Masahito Fujio
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Makoto Tsuboi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Huiting Bian
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Masashi Wakasugi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Hideharu Hibi
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| |
Collapse
|
12
|
Zhu M, Ding Q, Lin Z, Fu R, Zhang F, Li Z, Zhang M, Zhu Y. New Targets and Strategies for Rheumatoid Arthritis: From Signal Transduction to Epigenetic Aspect. Biomolecules 2023; 13:biom13050766. [PMID: 37238636 DOI: 10.3390/biom13050766] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that can lead to joint damage and even permanent disability, seriously affecting patients' quality of life. At present, the complete cure for RA is not achievable, only to relieve the symptoms to reduce the pain of patients. Factors such as environment, genes, and sex can induce RA. Presently, non-steroidal anti-inflammatory drugs, DRMADs, and glucocorticoids are commonly used in treating RA. In recent years, some biological agents have also been applied in clinical practice, but most have side effects. Therefore, finding new mechanisms and targets for treating RA is necessary. This review summarizes some potential targets discovered from the perspective of epigenetics and RA mechanisms.
Collapse
Affiliation(s)
- Menglin Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Qian Ding
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Zhongxiao Lin
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Rong Fu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Fuyuan Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Zhaoyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Mei Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, School of Pharmacy, Macau University of Science and Technology, Macau 999078, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
13
|
Pascalau NA, Radu AF, Cseppento DCN, Andronie-Cioara FL, Jurcau A, Mos C, Bungau AF, Bungau SG. An Evidence-Based Update on the Potential Association between Rheumatoid Arthritis and Lymphangioleiomyomatosis. J Pers Med 2023; 13:jpm13040607. [PMID: 37108993 PMCID: PMC10141996 DOI: 10.3390/jpm13040607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023] Open
Abstract
Lymphangioleiomyomatosis (LAM) represents an uncommon disorder characterized by cystic lung destruction and chronic respiratory failure. Lung damage caused by various mechanisms may represent a hypothesis for studying the association between LAM and rheumatoid arthritis (RA), which is the most prevalent autoinflammatory rheumatic disease and may affect the lungs as an extra-articular manifestation. Despite their distinct clinical presentations, the pathophysiology of both disorders includes dysregulated immunological function, abnormal cellular development, and inflammation. Current research suggests a potential relationship between RA and LAM, as some RA patients have been reported to develop LAM. However, the association of RA and LAM raises important therapeutic dilemmas. For this reason, the trajectory of a patient who was identified in our medical records as suffering from both LAM and RA, treated with many novel molecules and biological therapy, but with a negative outcome due to respiratory and multiorgan failure, has been exemplified. The delay in the diagnosis of LAM is due to a correlation between RA and LAM, worsening the vital prognosis and also hindering pulmonary transplantation. In addition, extensive research is essential for understanding the potential connection between these two disorders and discovering any similar mechanisms involved that may underlie their occurrence. This may contribute to the development of new therapeutic options that target shared pathways implicated in the pathogenesis of RA and LAM.
Collapse
|
14
|
Extra-Cellular Vesicles Derived from Thyroid Cancer Cells Promote the Epithelial to Mesenchymal Transition (EMT) and the Transfer of Malignant Phenotypes through Immune Mediated Mechanisms. Int J Mol Sci 2023; 24:ijms24032754. [PMID: 36769076 PMCID: PMC9917007 DOI: 10.3390/ijms24032754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Thyroid cancer is the most common endocrine cancer, and its incidence is increasing in many countries around the world. Among thyroid cancers, the papillary thyroid cancer (PTC) histotype is particularly prevalent. A small percentage of papillary tumors is associated with metastases and aggressive behavior due to de-differentiation obtained through the epithelial-mesenchymal transition (EMT) by which epithelial thyroid cells acquire a fibroblast-like morphology, reduce cellular adhesion, increase motility and expression of mesenchymal proteins. The tumor microenvironment plays an important role in promoting an aggressive phenotype through hypoxia and the secretion of HMGB1 and other factors. Hypoxia has been shown to drastically change the tumor cell phenotype and has been associated with increasing metastatic and migratory behavior. Cells transfer information to neighboring cells or distant locations by releasing extracellular membrane vesicles (EVs) that contain key molecules, such as mRNAs, microRNAs (miRNAs), and proteins, that are able to modify protein expression in recipient cells. In this study, we investigated the potential role of EVs released by the anaplastic cancer cell line CAL-62 in inducing a malignant phenotype in a papillary cancer cell line (BCPAP).
Collapse
|
15
|
Norda S, Papadantonaki R. Regulation of cells of the arterial wall by hypoxia and its role in the development of atherosclerosis. VASA 2023; 52:6-21. [PMID: 36484144 DOI: 10.1024/0301-1526/a001044] [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: 12/14/2022]
Abstract
The cell's response to hypoxia depends on stabilization of the hypoxia-inducible factor 1 complex and transactivation of nuclear factor kappa-B (NF-κB). HIF target gene transcription in cells resident to atherosclerotic lesions adjoins a complex interplay of cytokines and mediators of inflammation affecting cholesterol uptake, migration, and inflammation. Maladaptive activation of the HIF-pathway and transactivation of nuclear factor kappa-B causes monocytes to invade early atherosclerotic lesions, maintaining inflammation and aggravating a low-oxygen environment. Meanwhile HIF-dependent upregulation of the ATP-binding cassette transporter ABCA1 causes attenuation of cholesterol efflux and ultimately macrophages becoming foam cells. Hypoxia facilitates neovascularization by upregulation of vascular endothelial growth factor (VEGF) secreted by endothelial cells and vascular smooth muscle cells lining the arterial wall destabilizing the plaque. HIF-knockout animal models and inhibitor studies were able to show beneficial effects on atherogenesis by counteracting the HIF-pathway in the cell wall. In this review the authors elaborate on the up-to-date literature on regulation of cells of the arterial wall through activation of HIF-1α and its effect on atherosclerotic plaque formation.
Collapse
Affiliation(s)
- Stephen Norda
- Department of Cardiovascular Medicine, University Hospital Münster, Germany
| | - Rosa Papadantonaki
- Emergency Department, West Middlesex University Hospital, Chelsea and Westminster NHS Trust, London, United Kingdom
| |
Collapse
|
16
|
Peng Y, Li Y, Yang Y, Gao Y, Ren H, Hu J, Cui X, Lu W, Tao H, Chen Z. The genus Porana (Convolvulaceae) - A phytochemical and pharmacological review. Front Pharmacol 2022; 13:998965. [PMID: 36330088 PMCID: PMC9622789 DOI: 10.3389/fphar.2022.998965] [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: 07/20/2022] [Accepted: 10/10/2022] [Indexed: 11/27/2022] Open
Abstract
There are about 20 species of Porana Burm. f. worldwide in tropical and subtropical Asia, Africa and neighboring islands, Oceania, and the Americas. In China, India, and other places, this genus enjoys a wealth of experience in folk applications. Nevertheless, the chemical composition of only five species has been reported, and 59 compounds have been isolated and identified, including steroids, coumarins, flavonoids, quinic acid derivatives, and amides. Pharmacological studies revealed that extracts from this genus and their bioactive components exhibit anti-inflammatory, analgesic, antioxidant, anti-gout, anti-cancer, and anti-diabetic effects. Although this genus is abundant, the development of its pharmacological applications remains limited. This review will systematically summarize the traditional and current uses, chemical compositions, and pharmacological activities of various Porana species. Network analysis was introduced to compare and confirm its output with current research progress to explore the potential targets and pathways of chemical components in this genus. We hope to increase understanding of this genus’s medicinal value and suggest directions for rational medicinal development.
Collapse
Affiliation(s)
- Yu Peng
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ye Li
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Yuanyuan Yang
- Xi’an Institute for Food and Drug Control, Xi’an, Shaanxi, China
| | - Yuanqing Gao
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Hui Ren
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Jing Hu
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Xiaomin Cui
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Wenjing Lu
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
| | - Hongxun Tao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
- *Correspondence: Hongxun Tao, ; Zhiyong Chen,
| | - Zhiyong Chen
- Shaanxi Academy of Traditional Chinese Medicine, Xi’an, Shaanxi, China
- *Correspondence: Hongxun Tao, ; Zhiyong Chen,
| |
Collapse
|
17
|
Yue Q, Song Y, Liu Z, Zhang L, Yang L, Li J. Receptor for Advanced Glycation End Products (RAGE): A Pivotal Hub in Immune Diseases. Molecules 2022; 27:molecules27154922. [PMID: 35956875 PMCID: PMC9370360 DOI: 10.3390/molecules27154922] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/21/2022] [Accepted: 07/31/2022] [Indexed: 02/07/2023] Open
Abstract
As a critical molecule in the onset and sustainment of inflammatory response, the receptor for advanced glycation end products (RAGE) has a variety of ligands, such as advanced glycation end products (AGEs), S100/calcium granule protein, and high-mobility group protein 1 (HMGB1). Recently, an increasing number studies have shown that RAGE ligand binding can initiate the intracellular signal cascade, affect intracellular signal transduction, stimulate the release of cytokines, and play a vital role in the occurrence and development of immune-related diseases, such as systemic lupus erythematosus, rheumatoid arthritis, and Alzheimer’s disease. In addition, other RAGE signaling pathways can play crucial roles in life activities, such as inflammation, apoptosis, autophagy, and endoplasmic reticulum stress. Therefore, the strategy of targeted intervention in the RAGE signaling pathway may have significant therapeutic potential, attracting increasing attention. In this paper, through the systematic induction and analysis of RAGE-related signaling pathways and their regulatory mechanisms in immune-related diseases, we provide theoretical clues for the follow-up targeted intervention of RAGE-mediated diseases.
Collapse
Affiliation(s)
- Qing Yue
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (Q.Y.); (Y.S.); (Z.L.); (L.Y.)
| | - Yu Song
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (Q.Y.); (Y.S.); (Z.L.); (L.Y.)
| | - Zi Liu
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (Q.Y.); (Y.S.); (Z.L.); (L.Y.)
| | - Lin Zhang
- Department of Internal Medicine Nursing, School of Nursing, Wannan Medical College, 22 Wenchang West Road, Higher Education Park, Wuhu 241002, China;
| | - Ling Yang
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (Q.Y.); (Y.S.); (Z.L.); (L.Y.)
| | - Jinlong Li
- Hebei Key Laboratory for Organ Fibrosis Research, School of Public Health, North China University of Science and Technology, Tangshan 063210, China; (Q.Y.); (Y.S.); (Z.L.); (L.Y.)
- Correspondence: ; Tel.: +86-0315-8805572
| |
Collapse
|
18
|
Danieli MG, Antonelli E, Piga MA, Claudi I, Palmeri D, Tonacci A, Allegra A, Gangemi S. Alarmins in autoimmune diseases. Autoimmun Rev 2022; 21:103142. [PMID: 35853572 DOI: 10.1016/j.autrev.2022.103142] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/10/2022] [Indexed: 12/18/2022]
Abstract
Alarmins are endogenous, constitutively expressed, chemotacting and immune activating proteins or peptides released because of non-programmed cell death (i.e. infections, trauma, etc). They are considered endogenous damage-associated molecular patterns (DAMPs), able to induce a sterile inflammation. In the last years, several studies highlighted a possible role of different alarmins in the pathogenesis of various autoimmune and immune-mediated diseases. We reviewed the relevant literature about this topic, for about 160 articles. Particularly, we focused on systemic autoimmune diseases (systemic lupus erythematosus, rheumatoid arthritis, idiopathic inflammatory myopathies, ANCA-associated vasculitides, Behçet's disease) and cutaneous organ-specific autoimmune diseases (vitiligo, psoriasis, alopecia, pemphigo). Finally, we discussed about future perspectives and potential therapeutic implications of alarmins in autoimmune diseases. In fact, identification of receptors and downstream signal transducers of alarmins may lead to the identification of antagonistic inhibitors and agonists, with the capacity to modulate alarmins-related pathways and potential therapeutic applicability.
Collapse
Affiliation(s)
- Maria Giovanna Danieli
- Clinica Medica, Dipartimento di Scienze Cliniche e Molecolari, Università Politecnica delle Marche, via Tronto 10/A, 60126 Torrette di Ancona, Italy; Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Eleonora Antonelli
- PostGraduate School of Internal Medicine, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Mario Andrea Piga
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Ilaria Claudi
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Davide Palmeri
- Postgraduate School of Allergy and Clinical Immunology, Università Politecnica delle Marche, via Tronto 10/A, 60126 Ancona, Italy.
| | - Alessandro Tonacci
- Institute of Clinical Physiology, National Research Council of Italy (IFC-CNR), Via G. Moruzzi 1, 56124 Pisa, Italy.
| | - Alessandro Allegra
- Division of Haematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy.
| |
Collapse
|
19
|
Dong Y, Ming B, Dong L. The Role of HMGB1 in Rheumatic Diseases. Front Immunol 2022; 13:815257. [PMID: 35250993 PMCID: PMC8892237 DOI: 10.3389/fimmu.2022.815257] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/31/2022] [Indexed: 12/19/2022] Open
Abstract
HMGB1, a highly conserved non-histone nuclear protein, is widely expressed in mammalian cells. HMGB1 in the nucleus binds to the deoxyribonucleic acid (DNA) to regulate the structure of chromosomes and maintain the transcription, replication, DNA repair, and nucleosome assembly. HMGB1 is actively or passively released into the extracellular region during cells activation or necrosis. Extracellular HMGB1 as an alarmin can initiate immune response alone or combined with other substances such as nucleic acid to participate in multiple biological processes. It has been reported that HMGB1 is involved in various inflammatory responses and autoimmunity. This review article summarizes the physiological function of HMGB1, the post-translational modification of HMGB1, its interaction with different receptors, and its recent advances in rheumatic diseases and strategies for targeted therapy.
Collapse
Affiliation(s)
- Yuanji Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bingxia Ming
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Wang P, Hao P, Chen X, Li L, Zhou Y, Zhang X, Zhu L, Ying M, Han R, Wang L, Li X. Targeting HMGB1-NFκb Axis and miR-21 by Glycyrrhizin: Role in Amelioration of Corneal Injury in a Mouse Model of Alkali Burn. Front Pharmacol 2022; 13:841267. [PMID: 35586052 PMCID: PMC9108160 DOI: 10.3389/fphar.2022.841267] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/01/2022] [Indexed: 11/13/2022] Open
Abstract
Corneal neovascularization (CNV) is a sight-threatening condition usually associated with various inflammatory settings including chemical injury. High mobility group box 1 (HMGB1) is identified as an inflammatory alarmin in diverse tissue damage. Here, we evaluate the expression of HMGB1 and the consequences of its inhibition through its selective inhibitor glycyrrhizin (GLY) in alkali burn-induced corneal inflammation and neovascularization. GLY effectively attenuated alkali burn-induced HMGB1 expression at both mRNA and protein levels. Furthermore, slit-lamp analysis, ink perfusion, H&E staining, and CD31 histochemical staining showed that GLY relieved corneal neovascularization, while GLY attenuated VEGF expression via inhibiting HMGB1/NF-κB/HIF-1α signal pathway. In addition, GLY treatment decreased the cytokine expression of CCL2 and CXCL5, accompanied by the reduction of their receptors of CCR2 and CXCR2. GLY diminished the inflammatory cell infiltration of the cornea, as well as reduced the expression of IL-1β, IL-6, and TNF-α. Moreover, treatment with GLY reduced the degree of cornea opacity through inactivating extracellular HMGB1 function, which otherwise induces TGF-β1 release and myofibroblast differentiation. Furthermore, we found that GLY treatment attenuated the upregulation of miR-21 levels in alkali burned cornea; while inhibition of miR-21in keratocytes in vitro, significantly inhibited TGF-β1-induced myofibroblast differentiation. Collectively, our results suggested that targeting HMGB1-NFκb axis and miR-21 by GLY could introduce a therapeutic approach to counter CNV.
Collapse
Affiliation(s)
- Peihong 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
| | - 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 Affiliated Eye Hospital, Tianjin, China
| | - Xi Chen
- 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 Affiliated Eye Hospital, Tianjin, China
| | - Linghan 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
| | - 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
| | - Xiaohan Zhang
- 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
| | - Lin Zhu
- 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
| | - 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 Affiliated 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 Affiliated 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 Affiliated 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 Affiliated Eye Hospital, Tianjin, China
- *Correspondence: Xuan Li,
| |
Collapse
|
21
|
Hu J, Zhao L, Li N, Yang Y, Qu T, Ren H, Cui X, Tao H, Chen Z, Peng Y. Investigation of the active ingredients and pharmacological mechanisms of Porana sinensis Hemsl. Against rheumatoid arthritis using network pharmacology and experimental validation. PLoS One 2022; 17:e0264786. [PMID: 35235611 PMCID: PMC8890728 DOI: 10.1371/journal.pone.0264786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
Background Porana sinensis Hemsl. has been widely used as a substitute for Erycibes Caulis to treat rheumatoid arthritis (RA) in traditional Chinese medicine (TCM). However, little is known about the active ingredients and pharmacological mechanisms that mediate the action of P. sinensis against RA. Methods The compounds contained in P. sinensis were analyzed by Q Exactive Focus mass spectrometer. The active constituents and pharmacological mechanism of P. sinensis against RA were clarified using a network pharmacology-based investigation. LPS-induced RAW 264.7 cells was used to verify anti-inflammatory effects of the active compounds screened by network pharmacology. Collagen-induced arthritis model was used to further investigate the mechanism of P. sinensis against RA. Results The potential components and targets of P. sinensis against RA were analyzed using network pharmacology, and five compounds, twenty-five targets, and eight pathways were identified. Experimental validation suggested that P. sinensis extract and five compounds (esculetin, umbelliferone, trans-N-feruloyltyramine, caffeic acid and scopolin) could inhibit the release of inflammatory mediators (NO, TNF-α, IL-1β and IL-6) in LPS-induced RAW 264.7 cell. P. sinensis extract attenuated the severity, pathological changes, and release of cytokines (IL-6 and HIF-1α) during RA progression by regulating the PI3K/AKT and HIF-1 pathways. Conclusion The study provides a basis for the application of P. sinensis against RA. Our findings may provide suggestions for developing P. sinensis into a substitute for Erycibes Caulis.
Collapse
Affiliation(s)
- Jing Hu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
| | - Lintao Zhao
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
| | - Ning Li
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
- * E-mail: (NL); (ZC); (YP)
| | - Yuanyuan Yang
- Department of Traditional Chinese Medicine, Xi’an Institute for Food and Drug Control, Xi’an, China
| | - Tong Qu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
| | - Hui Ren
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
| | - Xiaomin Cui
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
| | - Hongxun Tao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Zhiyong Chen
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi’an, China
- * E-mail: (NL); (ZC); (YP)
| | - Yu Peng
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
- * E-mail: (NL); (ZC); (YP)
| |
Collapse
|
22
|
Targeting necroptosis in muscle fibers ameliorates inflammatory myopathies. Nat Commun 2022; 13:166. [PMID: 35013338 PMCID: PMC8748624 DOI: 10.1038/s41467-021-27875-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 12/16/2021] [Indexed: 12/12/2022] Open
Abstract
Muscle cell death in polymyositis is induced by CD8+ cytotoxic T lymphocytes. We hypothesized that the injured muscle fibers release pro-inflammatory molecules, which would further accelerate CD8+ cytotoxic T lymphocytes-induced muscle injury, and inhibition of the cell death of muscle fibers could be a novel therapeutic strategy to suppress both muscle injury and inflammation in polymyositis. Here, we show that the pattern of cell death of muscle fibers in polymyositis is FAS ligand-dependent necroptosis, while that of satellite cells and myoblasts is perforin 1/granzyme B-dependent apoptosis, using human muscle biopsy specimens of polymyositis patients and models of polymyositis in vitro and in vivo. Inhibition of necroptosis suppresses not only CD8+ cytotoxic T lymphocytes-induced cell death of myotubes but also the release of inflammatory molecules including HMGB1. Treatment with a necroptosis inhibitor or anti-HMGB1 antibodies ameliorates myositis-induced muscle weakness as well as muscle cell death and inflammation in the muscles. Thus, targeting necroptosis in muscle cells is a promising strategy for treating polymyositis providing an alternative to current therapies directed at leukocytes.
Collapse
|
23
|
Ji H, Zhang Q, Yang Z, Rui M, Chen Q, Niu X. Molecular Mechanism of Extractum Liquidum Drug Loading Materials on Promoting Chronic Wound Tissue Repair Through Phosphatidylinositol 3 Kinase/Protein Kinase B/Hypoxia Induction Factor 1 α Signal Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
To solve the shortcomings of traditional Zeji extractum liquidum (traditional Chinese medicine used for wound healing), and to explore the effect of Zeji Etractum Lquidum (ZLE) Nano Materials (ZLENM) on chronic wound (CW) healing and its molecular mechanism. 30 SD rats were divided
into 3 groups in random: control group (Ctrl group), model group (CW group), and treatment group (ZLENM group). The results of wound healing rate showed that, in contrast with the CW, the healing rate of back wounds in the ZLENM group was greatly increased on the 7th and 14th days (P
< 0.05). In contrast with the Ctrl, the rats in the CW and the ZLENM groups had greatly increased CD31 positive staining on the 7th and 14th days (P < 0.05), and the CW was lower than the ZLENM group (P < 0.05). In contrast with the 7th day, the MVD in the CW and the
ZLENM groups was greatly reduced on the 14th day (P < 0.05). Western blot analysis of the expression of related signal molecules showed that the expressions of P-Akt, P-PI3K, HIF-1α, and VEGFR2 protein in the wounds in the CW and ZLENM groups were greatly increased in
contrast with the Ctrl (P < 0.05), and CW was lower than ZLENM group (P < 0.05). In conclusion, ZLENM can promote wound healing and increase the number of wound angiogenesis in CW rats. The mechanism is related to the activation of phosphatidylinositol 3 kinase/protein
kinase B/hypoxia induction factor 1α (PI3K/AKT/HIF-1α) signaling pathway.
Collapse
Affiliation(s)
- Hui Ji
- Department of Orthopedic, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, 210014, Jiangsu, China
| | - Qian Zhang
- Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zengmin Yang
- Department of Orthopedic, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, 210014, Jiangsu, China
| | - Minjie Rui
- Department of Orthopedic, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, 210014, Jiangsu, China
| | - Qiyi Chen
- Department of Orthopedic, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, 210014, Jiangsu, China
| | - Xiaohong Niu
- Department of Luoli, Nanjing Integrated Traditional Chinese and Western Medicine Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, 210014, Jiangsu, China
| |
Collapse
|
24
|
Yang W, Wei X, Jiao Y, Bai Y, Sam WN, Yan Q, Sun X, Li G, Ma J, Wei W, Tian D, Zheng F. STAT3/HIF-1α/fascin-1 axis promotes RA FLSs migration and invasion ability under hypoxia. Mol Immunol 2021; 142:83-94. [PMID: 34971867 DOI: 10.1016/j.molimm.2021.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/15/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022]
Abstract
Rheumatoid arthritis (RA) synovium was identified as "tumor-like" tissues because of the hypoxic microenvironment, significant cell proliferation, and invasion phenotypes. It was reported that hypoxia promoted tumor aggressiveness via up-regulated expression of fascin-1 in cancer. However, the role of fascin-1 in RA synovial hyperplasia and joint injury progression remains unknown. In the current study, we first identified that both fascin-1 and HIF-1α were highly expressed in the RA synovium, in which they were widely colocalized, compared to osteoarthritis(OA). As well, levels of fascin-1 in RA fibroblast-like synoviocytes(FLSs) were found significantly higher than those in OA FLSs. Further, it was demonstrated that the mRNA and protein levels of fascin-1 in RA FLSs were up-regulated in hypoxia (3 % O2) and experimental hypoxia induced by cobalt chloride. Mechanistically, the HIF-1α-mediated hypoxia environment activated the gene expression of the fascin-1 protein, which in turn promoted the migration and invasion of RA FLSs. Accordingly, the restoration of FLSs migration and invasion was observed following siRNA-mediated silencing of fascin-1 and HIF-1α expression. Notably, under the experimental hypoxia, we found that the expression levels of fascin-1, HIF-1α, and p-STAT3 were increased in a time-dependent manner, and fascin-1and HIF-1α expressions were dependent on p-STAT3. Our results indicated that hypoxia-induced fascin-1 up-regulation promoted RA FLSs migration and invasion through the STAT3/HIF-1α/fascin-1 axis, which might represent a novel therapeutic target for the treatment of RA.
Collapse
Affiliation(s)
- Wang Yang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xinyue Wei
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Yachong Jiao
- Department of Clinical Laboratory, The Third Hospital of Hebei Medical University, Hebei, China
| | - Yingyu Bai
- Laboratory for Mechanisms and Therapies of Heart Diseases, School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Wilfried Noel Sam
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Qiushuang Yan
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China
| | - Guangping Li
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin, China.
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin, China.
| | - Derun Tian
- Department of Clinical Laboratory Diagnostics, Tianjin Medical University, Tianjin, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
25
|
Ao L, Gao H, Jia L, Liu S, Guo J, Liu B, Dong Q. Matrine inhibits synovial angiogenesis in collagen-induced arthritis rats by regulating HIF-VEGF-Ang and inhibiting the PI3K/Akt signaling pathway. Mol Immunol 2021; 141:13-20. [PMID: 34781187 DOI: 10.1016/j.molimm.2021.11.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 10/10/2021] [Accepted: 11/05/2021] [Indexed: 12/19/2022]
Abstract
Matrine (Mat) is an alkaloid of tetracycline quinazine, and previous studies have demonstrated its specific effect on relieving rheumatoid arthritis (RA). However, the effect of Mat on joint synovial angiogenesis in the pathogenesis of RA has not been elucidated. In this study, body weight, joint swelling, arthritis index (AI) score, histopathological changes, immunohistochemical, and western blot- were used in collagen-induced arthritis (CIA) rats to detect pro-inflammatory factors and, - expression levels of key cytokines and proteins along the hypoxia-inducible factor (HIF)-endothelial growth factor (VEGF)-angiopoietin (Ang) axis and VEGF-phosphoinositide 3-kinase (PI3K) / protein kinase B (Akt) pathway. In vitro experiments were conducted to observe the effect of Mat on the proliferation, migration and lumen formation of RA-fibroblast-like synovial cells (FLS) and human umbilical vein endothelial cells (HUVECs). Results showed that Mat reduced the degree of paw swelling and AI score in CIA rats, joint synovial tissue proliferation, inflammatory cell infiltration, and neovascularization; moreover, it down-regulated the expression levels of inflammatory factors interleukin-1β, interferon-γ, and pro-angiogenic factors VEGF, placental growth factor, HIF-α, Ang-1, Ang-2, Tie-2, and phosphorylation-Akt in the ankle joint of CIA rats. In addition, the in vitro experiments showed that Mat inhibited the proliferation and migration of RA-FLS and inhibited the proliferation and lumen formation of HUVECs. Therefore, Mat exerts an anti-angiogenesis effect by regulating the HIF-VEGF-Ang axis and inhibiting the PI3K/Akt signaling pathway. This inhibits the pathogenesis and improve the symptoms of RA, and may be offered as a candidate drug for the treatment of RA.
Collapse
Affiliation(s)
- Limei Ao
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Han Gao
- Department of Rheumatology and Immunology, Chifeng Hospital of Mongolian Medicine and Traditional Chinese Medicine, Chifeng, 024000, China
| | - Lifen Jia
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Shimin Liu
- Department of Urology, Inner Mongolia Autonomous Region Hospital of Traditional Chinese Medicine, Huhhot, 010110, China
| | - Jie Guo
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Bingzhen Liu
- Department of TCM Rheumatology, Huhhot Hospital of Mongolian Medicine and Traditional Chinese Medicine, Huhhot, 010110, China
| | - Qiumei Dong
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Huhhot, 010110, China.
| |
Collapse
|
26
|
Wang Y, Wu H, Deng R. Angiogenesis as a potential treatment strategy for rheumatoid arthritis. Eur J Pharmacol 2021; 910:174500. [PMID: 34509462 DOI: 10.1016/j.ejphar.2021.174500] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 08/04/2021] [Accepted: 09/06/2021] [Indexed: 12/15/2022]
Abstract
Angiogenesis is an early and key event in the pathogenesis of rheumatoid arthritis (RA) and is crucial for the proliferation of synovial tissue and the formation of pannus. This process is regulated by both angiogenesis-stimulating factors and angiogenesis inhibitors, the basis for the "on-off hypothesis of angiogenesis." In RA, inflammation, immune imbalance, and hypoxia can further turn on the switch for blood vessel formation and induce angiogenesis. The new vasculature can recruit white blood cells, induce immune imbalance, and aggravate inflammation. At the same time, it also can provide oxygen and nutrients for the proliferating synovial tissue, which can accelerate the process of RA. The current therapies for RA mainly target the inflammatory response of autoimmune activation. Although these therapies have been greatly improved, there are still many patients whose RA is difficult to treat or who do not fully respond to treatment. Therefore, new innovative therapies are still urgently needed. This review covers the mechanism of synovial angiogenesis in RA, including the detailed process of angiogenesis and the relationship between inflammation, immune imbalance, hypoxia, and synovial angiogenesis, respectively. At the same time, in the context of the development of angiogenesis inhibition therapy for cancer, we also discuss similar treatment strategies for RA, especially the combination of targeted angiogenesis inhibition therapy and immunotherapy.
Collapse
Affiliation(s)
- Yan Wang
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| | - Hong Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China.
| | - Ran Deng
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China; Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei, 230012, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, 230012, China
| |
Collapse
|
27
|
Amaral-Silva D, Gonçalves R, Torrão RC, Torres R, Falcão S, Gonçalves MJ, Araújo MP, Martins MJ, Lopes C, Neto A, Marona J, Costa T, Castelão W, Silva AB, Silva I, Lourenço MH, Mateus M, Gonçalves NP, Manica S, Costa M, Pimentel-Santos FM, Mourão AF, Branco JC, Soares H. Direct tissue-sensing reprograms TLR4 + Tfh-like cells inflammatory profile in the joints of rheumatoid arthritis patients. Commun Biol 2021; 4:1135. [PMID: 34580414 PMCID: PMC8476501 DOI: 10.1038/s42003-021-02659-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
CD4+ T cells mediate rheumatoid arthritis (RA) pathogenesis through both antibody-dependent and independent mechanisms. It remains unclear how synovial microenvironment impinges on CD4+ T cells pathogenic functions. Here, we identified a TLR4+ follicular helper T (Tfh) cell-like population present in the blood and expanded in synovial fluid. TLR4+ T cells possess a two-pronged pathogenic activity whereby direct TLR4+ engagement by endogenous ligands in the arthritic joint reprograms them from an IL-21 response, known to sponsor antibody production towards an IL-17 inflammatory program recognized to fuel tissue damage. Ex vivo, synovial fluid TLR4+ T cells produced IL-17, but not IL-21. Blocking TLR4 signaling with a specific inhibitor impaired IL-17 production in response to synovial fluid recognition. Mechanistically, we unveiled that T-cell HLA-DR regulates their TLR4 expression. TLR4+ T cells appear to uniquely reconcile an ability to promote systemic antibody production with a local synovial driven tissue damage program. In order to identify how the synovial microenvironment impinges on CD4+ T cells pathogenic functions in Rheumatoid Arthritis (RA), Amaral-Silva examined RA patient blood and synovial fluif and identified the presence of a TLR4+ follicular helper T (Tfh) cell-like population. They provided mechanistic insight into how TLR4+ T cells uniquely reconcile an ability to promote systemic antibody production with a local synovial driven-tissue damage program.
Collapse
Affiliation(s)
- Daniela Amaral-Silva
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rute Gonçalves
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rita C. Torrão
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Rita Torres
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Sandra Falcão
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Maria João Gonçalves
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria Paula Araújo
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria José Martins
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Carina Lopes
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Agna Neto
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - José Marona
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Tiago Costa
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Walter Castelão
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Ana Bento Silva
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Inês Silva
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Maria Helena Lourenço
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Margarida Mateus
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Nuno Pina Gonçalves
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Santiago Manica
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Manuela Costa
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
| | - Fernando M. Pimentel-Santos
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Ana Filipa Mourão
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
| | - Jaime C. Branco
- grid.414462.10000 0001 1009 677XHospital Egas Moniz, Rua da Junqueira n° 126, Lisboa, Portugal
- Rheumatological Diseases Laboratory, Lisboa, Portugal
- grid.10772.330000000121511713CHRC|CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Group, Lisboa, Portugal
- grid.10772.330000000121511713iNOVA4Health | CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisboa, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Lisbon Campus, Rua do Instituto Bacteriológico 5, Lisboa, Portugal
| |
Collapse
|
28
|
Oda H, Nagamatsu T, Cabral H, Miyazaki T, Iriyama T, Kawana K, Fujii T, Osuga Y. Thrombomodulin promotes placental function by up-regulating placental growth factor via inhibition of high-mobility-group box 1 and hypoxia-inducible factor 1α. Placenta 2021; 111:1-9. [PMID: 34126415 DOI: 10.1016/j.placenta.2021.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/13/2021] [Accepted: 06/01/2021] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Pregnancy is a state of maternal systemic stress due to inflammation and hypoxic reactions originating from the utero-placental unit. Maternal tolerance to these stresses is a key for successful outcomes. Thrombomodulin (TM), a glycoprotein expressed on cell surface, regulates local inflammatory pathways by inhibiting proinflammatory factor, High-mobility-group box1(HMGB1). Although TM is highly expressed on placental trophoblast cells, biological activities of TM during pregnancy remains unclear. Here, we hypothesized that TM may contribute to the maternal stress coping mechanisms. METHODS By administering recombinant-TM (rTM) to the pregnant mice, we investigated the influence of TM functions on the placenta and fetal growth. We further examined its effect on trophoblast cells, focusing on HMGB1-regulated inflammatory signalings and hypoxia-inducible factor 1α (HIF1α)-dependent regulation of placental angiogenic factors. RESULTS Administration of rTM increased fetal weight and fetal/placental-weight ratios, which implies the improvement of placental function. These features were accompanied by maternal serum HMGB1 reduction and suppressed placental proinflammatory cytokine, IL-6 and TNF-α, expressions. In addition, rTM reduced HIF1α protein accumulation and enhanced placental growth factor (PlGF) expression in the placenta, that explains the improvement of maternal features. DISCUSSION Our study revealed the supportive effect of TM on the placental function in mice. By inhibiting HMGB1, rTM suppresses proinflammatory cytokines, downregulates HIF1α and induces PlFG expression in the placental tissue. Our results have elucidated the novel aspects of TM; the regulation of placental inflammatory cytokines and angiogenic factors, during pregnancy. These findings may reveal potential therapeutic opportunities for the management of maternal complications.
Collapse
Affiliation(s)
- Hiroko Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takuya Miyazaki
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Faculty of Medicine, Nihon University, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
He W, Xu F, Chen L, Huang W, Jiang L, Tang F, Yan W, Zhong S, Shen C, Huang H, Lv J, Wu X, Zeng S, Li M, Zhang M. Association of High-Mobility Group Box-1 with Inflammationrelated Cytokines in the Aqueous Humor with Acute Primary Angle-Closure Eyes. Curr Mol Med 2021; 21:237-245. [PMID: 32282301 DOI: 10.2174/1566524020666200413113107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/16/2020] [Accepted: 03/24/2020] [Indexed: 11/22/2022]
Abstract
AIM The aim of this study was to measure the levels of High-mobility group box-1 (HMGB1) and inflammation-related cytokines in the aqueous humor of patients with acute primary angle-closure glaucoma (APAG) and age-related cataract eyes (ARC). METHODS Aqueous humor samples were obtained from 59 eyes of 59 Chinese subjects (APAG, 32 eyes; and ARC, 27eyes). The multiplex bead immunoassay technique was used to measure the levels of HMGB1 and IL-8, IL-6, G-CSF, MCP-3, VEGF, sVEGFR- 1, sVEFGR-2, TNF-α, PDGF, and IL-10 in aqueous. The data of Patients' demographics and preoperative intraocular pressure (IOP) were also collected for detailed analysis. RESULTS The APAG group showed significantly elevated concentrations of HMGB1, IL- 8, IL-6, G-CSF, VEGF, sVEGFR-1, and TNF-α than those in the ARC group. Aqueous HMGB1 level correlated significantly with IOP, IL-8, IL-6, G-CSF and sVEGFR-1 levels but not with age, TNF-α, or VEGF levels. CONCLUSION The aqueous level of HMGB1 is elevated in APAG and associated with aqueous level of inflammation-related cytokines, suggesting an association between elevated levels of HMGB1, APAC and certain inflammatory modulators which, of course, should lead to further investigations in order to demonstrate the cause and effect.
Collapse
Affiliation(s)
- Wenjing He
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Fan Xu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Lifei Chen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Wei Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Li Jiang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Fen Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Wenya Yan
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shan Zhong
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Chaolan Shen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Hui Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Jian Lv
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Xiaonian Wu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Siming Zeng
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Min Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
30
|
Chen H, Lin X, Liu H, Huang C, Li R, Ai J, Wei J, Xiao S. HMGB1 Translocation is Associated with Tumor-Associated Myeloid Cells and Involved in the Progression of Fibroblastic Sarcoma. Pathol Oncol Res 2021; 27:608582. [PMID: 34257571 PMCID: PMC8262203 DOI: 10.3389/pore.2021.608582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/26/2021] [Indexed: 11/13/2022]
Abstract
The morphological variability and genetic complexity of fibroblastic sarcoma makes its diagnosis and treatment a challenge. High-mobility group box 1 protein (HMGB1), which functions as a DNA chaperone and a prototypical damage-associated molecular pattern, plays a paradoxical role in cancer. However, the expression pattern and role of HMGB1 in fibroblastic sarcomas is ill defined. By immunostaining of 95 tissue microarray cores of fibroblastic sarcomas, HMGB1 was found to be expressed in most tumor tissues. Nuclear HMGB1 translocation to cytoplasm was observed both in tumor cells and vascular endothelial cells. A visible number of tumor-associated myeloid cells including CD68+ and CD163+ macrophages and CD33+ myeloid cells were also detected in most tumor tissues. HMGB1 translocation was not only associated with CD68, CD163, and CD33 density, but also with disease progression. These results imply that HMGB1, an important regulator of the tumor microenvironment, is associated with tumor-associated myeloid cells and involved in the progression of fibroblastic sarcomas; HMGB1 may serve as a promising prognostic biomarker and a potential therapeutic target for fibroblastic sarcoma.
Collapse
Affiliation(s)
- Huoying Chen
- Prenatal Diagnosis Center, Guangdong Second Provincial General Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China.,Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Xiaoying Lin
- Prenatal Diagnosis Center, Guangdong Second Provincial General Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China
| | - Hongbo Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Cheng Huang
- Prenatal Diagnosis Center, Guangdong Second Provincial General Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China
| | - Rong Li
- Prenatal Diagnosis Center, Guangdong Second Provincial General Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China
| | - Jie Ai
- Department of Laboratory Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Jiaxue Wei
- Prenatal Diagnosis Center, Guangdong Second Provincial General Hospital, Guangdong Provincial Emergency Hospital, Guangzhou, China
| | - Shengjun Xiao
- Department of Pathology, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
31
|
Gualdoni GS, Jacobo PV, Sobarzo CM, Pérez CV, Durand LAH, Theas MS, Lustig L, Guazzone VA. Relevance of angiogenesis in autoimmune testis inflammation. Mol Hum Reprod 2021; 27:gaaa073. [PMID: 33313783 DOI: 10.1093/molehr/gaaa073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 10/01/2020] [Indexed: 12/27/2022] Open
Abstract
Experimental autoimmune orchitis (EAO) is a useful model to study organ-specific autoimmunity and chronic testicular inflammation. This model reflects testicular pathological changes reported in immunological infertility in men. Progression of EAO in rodents is associated with a significantly increased percentage of testicular endothelial cells and interstitial testicular blood vessels, indicating an ongoing angiogenic process. Vascular endothelial growth factor A (VEGFA), the main regulator of physiological and pathological angiogenesis, can stimulate endothelial cell proliferation, chemotaxis and vascular permeability. The aim of this study was to explore the role of VEGFA in the pathogenesis of testicular inflammation. Our results found VEGFA expression in Leydig cells, endothelial cells and macrophages in testis of rats with autoimmune orchitis. VEGFA level was significantly higher in testicular fluid and serum of rats at the end of the immunization period, preceding testicular damage. VEGF receptor (VEGFR) 1 is expressed mainly in testicular endothelial cells, whereas VEGFR2 was detected in germ cells and vascular smooth muscle cells. Both receptors were expressed in testicular interstitial cells. VEGFR2 increased after the immunization period in the testicular interstitium and VEGFR1 was downregulated in EAO testis. In-vivo-specific VEGFA inhibition by Bevacizumab prevented the increase in blood vessel number and reduced EAO incidence and severity. Our results unveil relevance of VEGFA-VEGFR axis during orchitis development, suggesting that VEGFA might be an early marker of testicular inflammation and Bevacizumab a therapeutic tool for treatment of testicular inflammation associated with subfertility and infertility.
Collapse
Affiliation(s)
- Gisela Soledad Gualdoni
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Patricia Verónica Jacobo
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Cristian Marcelo Sobarzo
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Cecilia Valeria Pérez
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Luis Alberto Haro Durand
- Instituto de Biología y Medicina Experimental (IBYME), CONICET, Ciudad Autónoma de Buenos Aires C1428ADN, Argentina
| | - María Susana Theas
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Livia Lustig
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| | - Vanesa Anabella Guazzone
- Departamento de Biología Celular e Histología/Unidad Académica II, Facultad de Medicina, Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED), Facultad de Medicina, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires (UBA), Ciudad Autónoma de Buenos Aires C1121ABG, Argentina
| |
Collapse
|
32
|
Kamiya N, Kim HKW. Elevation of Proinflammatory Cytokine HMGB1 in the Synovial Fluid of Patients With Legg-Calvé-Perthes Disease and Correlation With IL-6. JBMR Plus 2021; 5:e10429. [PMID: 33615102 PMCID: PMC7872337 DOI: 10.1002/jbm4.10429] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 10/16/2020] [Accepted: 10/31/2020] [Indexed: 12/22/2022] Open
Abstract
Legg-Calvé-Perthes disease (LCPD) is a childhood ischemic osteonecrosis (ON) of the femoral head associated with the elevation of proinflammatory cytokine interleukin-6 (IL-6) in the synovial fluid. Currently, there is no effective medical therapy for patients with LCPD. In animal models of ischemic ON, articular chondrocytes produce IL-6 in response to ischemic ON induction and IL-6 receptor blockade improves bone healing. High-mobility group box 1 (HMGB1) is a damage-associated molecular pattern released from dying cells. In addition, extracellular HMGB1 protein is a well-known proinflammatory cytokine elevated in the synovial fluid of patients with rheumatoid arthritis and osteoarthritis. The purpose of this study was to investigate IL-6-related proinflammatory cytokines, including HMGB1, in the synovial fluid of patients with LCPD. Our working hypothesis was that HMGB1, produced by articular chondrocytes following ischemic ON, plays an important role in IL-6 upregulation. Here, HMGB1 protein levels were significantly higher in the synovial fluid of patients with LCPD by threefold compared with controls (p < 0.05), and were highly correlated with IL-6 levels (Pearson correlation coefficient 0.94, p < 0.001, R 2 = 0.87). In the mouse model of ischemic ON, both HMGB1 gene expression and protein levels were elevated in the articular cartilage. In vitro studies revealed a significant elevation of HMGB1 and IL-6 proteins in the supernatants of human chondrocytes exposed to hypoxic and oxidative stresses. Overexpressed HMGB1 protein in the supernatants of chondrocytes synergistically increased IL-6 protein. Silencing HMGB1 RNA in human chondrocytes significantly repressed inteleukin-1β (IL-1β) gene expression, but not IL-6. Further, both IL-1β and tumor necrosis factor-α (TNF-α) protein levels in the synovial fluid of patients with LCPD were significantly correlated with IL-6 protein levels. Taken together, these results suggest that proinflammatory cytokines, HMGB1, tumor necrosis factor-α (TNF-α), and IL-1β, are significantly involved with IL-6 in the pathogenesis of LCPD. This study is clinically relevant because the availability of multiple therapeutic targets may improve the development of therapeutic strategy for LCPD. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nobuhiro Kamiya
- Center for Excellence in HipScottish Rite for ChildrenDallasTXUSA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Faculty of Budo and Sport StudiesTenri UniversityNaraJapan
| | - Harry KW Kim
- Center for Excellence in HipScottish Rite for ChildrenDallasTXUSA
- Department of Orthopedic SurgeryUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
33
|
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
|
34
|
Liu Z, Liu L, Cheng X, Gao L. Expression and predictive value of HIF-1α and VEGF in patients with burns following treatment. Exp Ther Med 2020; 20:141. [PMID: 33093879 PMCID: PMC7571334 DOI: 10.3892/etm.2020.9270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/15/2020] [Indexed: 11/28/2022] Open
Abstract
The present study aimed to investigate the expression and predictive value of serum hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) in patients with burns following treatment. A total of 84 patients with burns treated in Jinan City People's Hospital (Jinan, China) between June 2015 and August 2017 were selected and their clinical information was collected. The expression levels of HIF-1α and VEGF before and after treatment were detected via ELISA, and HIF-1α and VEGF levels in patients with effective and ineffective treatment were compared. The predictive values of HIF-1α and VEGF in clinical efficacy were determined using receiver operating characteristic (ROC) curves, and independent risk factors affecting treatment inefficacy were analyzed via multivariate logistic regression. It was revealed that HIF-1α decreased significantly (P<0.05) while VEGF significantly increased in patients after treatment. Patients with effective treatment presented significantly lower HIF-1α levels and higher VEGF levels compared with those with ineffective treatment. The ROC curve indicated that the area under the curve (AUC) of HIF-1α for treatment efficacy was 0.795, the 95% CI was 0.666-0.924, the specificity and sensitivity were 68.75 and 80.88%, respectively, and the Youden index was 49.63%. For VEGF, the AUC, 95% CI, specificity, sensitivity and Youden index were 0.826, 0.725-0.928, 68.75, 82.35 and 51.10% respectively. Moreover, under the joint detection of HIF-1α and VEGF, the AUC was 0.847, 95% CI was 0.746-0.947, specificity and sensitivity were 87.50 and 66.18%, respectively, with a Youden index of 53.68%. Multivariate analysis demonstrated that higher HIF-1α level, lower VEGF level and higher burn degree before treatment were independent risk factors for treatment inefficacy. HIF-1α levels decreased and VEGF levels increased in burn patients after treatment. HIF-1α and VEGF before treatment may therefore serve as predictors for treatment efficacy.
Collapse
Affiliation(s)
- Zhufeng Liu
- Department of Burn Dermatology, Jinan City People's Hospital, Jinan, Shandong 271100, P.R. China
| | - Ling Liu
- Department of Burn Dermatology, Jinan City People's Hospital, Jinan, Shandong 271100, P.R. China
| | - Xuejuan Cheng
- Department of Nursing, The People's Hospital of Zouping City, Binzhou, Shandong 256200, P.R. China
| | - Liming Gao
- Department of Burn Dermatology, Jinan City People's Hospital, Jinan, Shandong 271100, P.R. China
| |
Collapse
|
35
|
Xue J, Suarez JS, Minaai M, Li S, Gaudino G, Pass HI, Carbone M, Yang H. HMGB1 as a therapeutic target in disease. J Cell Physiol 2020; 236:3406-3419. [PMID: 33107103 DOI: 10.1002/jcp.30125] [Citation(s) in RCA: 186] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/13/2020] [Indexed: 12/30/2022]
Abstract
High-mobility group box 1 (HMGB1) was initially recognized as a ubiquitous nuclear protein involved in maintaining the nucleosome integrity and facilitating gene transcription. HMGB1 has since been reevaluated to be a prototypical damage-associated molecular pattern (DAMP) protein, and together with its exogenous counterpart, pathogen-associated molecular pattern (PAMP), completes the body's alarmin system against disturbances in homeostasis. HMGB1 can be released into the extracellular matrix (ECM) by either granulocytes or necrotic cells to serve as a chemotaxis/cytokine during infection, endotoxemia, hypoxia, ischemia-reperfusion events, and cancer. Different isoforms of HMGB1 present with distinctive physiological functions in ECM-fully-reduced HMGB1 (all thiol) acts as the initial damage signal to recruit circulating myeloid cells, disulfide HMGB1 behaves as a cytokine to activate macrophages and neutrophils, and both signals are turned off when HMGB1 is terminally oxidized into the final sulfonate form. Targeting HMGB1 constitutes a favorable therapeutic strategy for inflammation and inflammatory diseases. Antagonists such as ethyl pyruvate inhibit HMGB1 by interfering with its cytoplasmic exportation, while others such as glycyrrhizin directly bind to HMGB1 and render it unavailable for its receptors. The fact that a mixture of different HMGB1 isoforms is present in the ECM poses a challenge in pinpointing the exact role of an individual antagonist. A more discriminative probe for HMGB1 may be necessary to advance our knowledge of HMGB1, HMGB1 antagonists, and inflammatory-related diseases.
Collapse
Affiliation(s)
- Jiaming Xue
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA.,John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Joelle S Suarez
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Michael Minaai
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Shuangjing Li
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA.,Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Giovanni Gaudino
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York, USA
| | - Michele Carbone
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| | - Haining Yang
- Thoracic Oncology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, USA
| |
Collapse
|
36
|
Etanercept as a TNF-alpha inhibitor depresses experimental retinal neovascularization. Graefes Arch Clin Exp Ophthalmol 2020; 259:661-671. [PMID: 33043386 DOI: 10.1007/s00417-020-04956-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/07/2020] [Accepted: 10/01/2020] [Indexed: 01/06/2023] Open
Abstract
PURPOSE The formation of retinal neovascularization (RNV) is the primary pathological process underlying retinopathy of prematurity (ROP). Previous studies have shown that inflammatory factors are related to the formation of RNV. Tumor necrosis factor-α (TNF-α), as an important factor in the inflammatory response, is involved in the regulation of RNV formation. However, the mechanism through which TNF-α inhibition reduces RNV formation is not fully clarified. Therefore, the purpose of this study was to explore the effect of etanercept, an inhibitor of TNF-α, on RNV, and its possible mechanism. METHODS In vivo, an oxygen-induced retinopathy (OIR) mouse model was used to determine the effect of etanercept on the formation of RNV by performing immunostaining. The effect of etanercept on tumor necrosis factor receptor-associated factor 2 (TRAF2), pro-angiogenic-related factors, and pro/anti-inflammatory factors in OIR mice was assessed by real-time PCR and Western blotting. In vitro, the effect of etanercept on TNF-α-induced human retinal microvascular endothelial cell tube formation was evaluated by tube formation assays, and the potential mechanism of etanercept was explored by Western blotting. RESULTS In vivo, etanercept reduced the area of RNV and decreased the expression of TRAF2 in the OIR mouse model. Etanercept also suppressed the expression of several pro-angiogenic factors and regulated the pro/anti-inflammatory factors. In vitro, etanercept reduced endothelial cell tube formation by inhibiting activation of the NF-κB signaling pathway. CONCLUSION Etanercept can regulate pro/anti-inflammatory factors and reduce the expression of pro-angiogenic factors by inhibiting NF-κB phosphorylation, thereby reducing RNV formation.
Collapse
|
37
|
Polyakova YV, Zavodovsky BV, Sivordova LE, Akhverdyan YR, Zborovskaya IA. Visfatin and Rheumatoid Arthritis: Pathogenetic Implications and Clinical Utility. Curr Rheumatol Rev 2020; 16:224-239. [DOI: 10.2174/1573397115666190409112621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/15/2019] [Accepted: 04/02/2019] [Indexed: 12/12/2022]
Abstract
Objective:
Analysis and generalization of data related to visfatin involvement in the
pathogenesis of inflammation at various stages of rheumatoid arthritis.
Data Synthesis:
Visfatin is an adipocytokine which has also been identified in non-adipose tissues.
It influences directly on the maturation of B cells, which are involved in autoantibody production
and T cell activation. Visfatin can promote inflammation via regulation of pro-inflammatory cytokines
including TNF, IL-1β and IL-6. The concentration of circulating visfatin in rheumatoid arthritis
patients is higher compared to healthy individuals. Several studies suggest that visfatin level is
associated with rheumatoid arthritis activity, and its elevation may precede clinical signs of the relapse.
In murine collagen-induced arthritis, visfatin levels were also found to be elevated both in
inflamed synovial cells and in joint vasculature. Visfatin blockers have been shown to confer fast
and long-term attenuation of pathological processes; however, most of their effects are transient.
Other factors responsible for hyperactivation of the immune system can participate in this process
at a later stage. Treatment of rheumatoid arthritis with a combination of these blockers and inhibitors
of other mediators of inflammation can potentially improve treatment outcomes compared to
current therapeutic strategies. Recent advances in the treatment of experimental arthritis in mice as
well as the application of emerging treatment strategies obtained from oncology for rheumatoid arthritis
management could be a source of novel adipokine-mediated anti-rheumatic drugs.
Conclusion:
The ongoing surge of interest in anticytokine therapy makes further study of visfatin
highly relevant as it may serve as a base for innovational RA treatment.
Collapse
Affiliation(s)
- Yulia V. Polyakova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Boris V. Zavodovsky
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Larisa E. Sivordova
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Yuri R. Akhverdyan
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| | - Irina A. Zborovskaya
- Research Institute for Clinical and Experimental Rheumatology, Volgograd, Russian Federation
| |
Collapse
|
38
|
Jiang T, Kong B, Yan W, Wu C, Jiang M, Xu X, Xi X. Network Pharmacology to Identify the Pharmacological Mechanisms of a Traditional Chinese Medicine Derived from Trachelospermum jasminoides in Patients with Rheumatoid Arthritis. Med Sci Monit 2020; 26:e922639. [PMID: 32840241 PMCID: PMC7466841 DOI: 10.12659/msm.922639] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study used a network pharmacology approach to identify the pharmacological mechanisms of a traditional Chinese medicine derived from Trachelospermum jasminoides (Lindl.) Lem. in patients with rheumatoid arthritis (RA). MATERIAL AND METHODS Known compounds of T. jasminoides were obtained from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) database, the Shanghai Institute of Organic Chemistry of Chinese Academy of Science, Chemistry (CASC) database, and a literature search. Putative targets of identified compounds were predicted by SwissTargetPrediction. RA-related targets were achieved from the Therapeutic Target database, Drugbank database, Pharmacogenomics Knowledgebase, and Online Mendelian Inheritance in Man database. The protein-protein interaction (PPI) network was built by STRING. CluGO was utilized for Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analysis. RESULTS A total of 354 potential targets were predicted for the 17 bioactive compounds in T. jasminoides; 69 of these targets overlapped with RA-related targets. A PPI network was composed and 2 clusters of 59 and 42 nodes each were excavated. GO and KEGG enrichment analysis of the overlapping targets and the 2 clusters was mainly grouped into immunity, inflammation, estrogen, anxiety, and depression processes. CONCLUSIONS Our study illustrated that T. jasminoides alleviates RA through the interleukin-17 signaling pathway, the tumor necrosis factor signaling pathway, and other immune and inflammatory-related processes. It also may exert effects in regulating cell differentiation and potentially has anti-anxiety, anti-depression, and estrogen-like effects.
Collapse
Affiliation(s)
- Tao Jiang
- Department of Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland).,Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Bo Kong
- Department of Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Wei Yan
- Department of Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Changgui Wu
- Department of Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Min Jiang
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Xing Xu
- Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| | - Xiaobing Xi
- Department of Traumatology, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland).,Shanghai Key Laboratory for Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (mainland)
| |
Collapse
|
39
|
Metabolic reprogramming as a key regulator in the pathogenesis of rheumatoid arthritis. Inflamm Res 2020; 69:1087-1101. [DOI: 10.1007/s00011-020-01391-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/02/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
|
40
|
Kaur I, Behl T, Bungau S, Kumar A, Mehta V, Setia D, Uddin MS, Zengin G, Aleya L, Arora S. Exploring the therapeutic promise of targeting HMGB1 in rheumatoid arthritis. Life Sci 2020; 258:118164. [PMID: 32739467 DOI: 10.1016/j.lfs.2020.118164] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/25/2020] [Accepted: 07/25/2020] [Indexed: 12/22/2022]
Abstract
High mobility group box-1 (HMGB1) protein is a diverse, single polypeptide moiety, present in mammalian eukaryotic cells. In response to stimuli, this nuclear protein is actively secreted in to the extracellular compartment or passively released by the necrotic cells, in order to mediate inflammatory responses, by forming complexes with IL-1α, IL-1β, LPS and other moieties, and binding to RAGE, TLR and other receptor ligands, initiating downstream, signaling processes. This molecule acts as a proinflammatory cytokine and contributes to the progression of diseases like, acute lung injury, autoimmune liver damage, graft rejection immune response and arthritis. Small concentrations of HMGB1 are released during apoptosis, which facilitates oxidative regulation on Cys106, and propagates immune inactivating tolerogenic signals in the body. The review portrays the role of HMGB1 in rheumatoid arthritis, evidently supported by pre-clinical and clinical investigations, demonstrating extensive HMGB1 expression in synovial tissue and fluid as well as serum, excessive expression of transduction receptor signaling molecules, bone remodeling and uncontrolled expression of bone destroying osteoclastogenesis, resulting in destruction of articular cartilage, bone deformation and synovial proliferation, alleviating the pathogenesis in RA disease. Moreover, the review highlights the therapeutic regime targeting HMGB1, facilitating inhibition of its actions and release into the extracellular compartment, to ameliorate the destructive events that prevail in rheumatoid arthritis.
Collapse
Affiliation(s)
- Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine of Pharmacy, University of Oradea, Oradea, Romania
| | - Arun Kumar
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Rohru, Distt. Shimla, Himachal Pradesh, India
| | - Dhruv Setia
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey
| | - Lotfi Aleya
- Department of Biology, Faculty of Science, University Campus, Konya, Turkey; Chrono-Environment Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, France
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
41
|
Nebbioso M, Lambiase A, Armentano M, Tucciarone G, Bonfiglio V, Plateroti R, Alisi L. The Complex Relationship between Diabetic Retinopathy and High-Mobility Group Box: A Review of Molecular Pathways and Therapeutic Strategies. Antioxidants (Basel) 2020; 9:antiox9080666. [PMID: 32722545 PMCID: PMC7464385 DOI: 10.3390/antiox9080666] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/06/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a protein that is part of a larger family of non-histone nuclear proteins. HMGB1 is a ubiquitary protein with different isoforms, linked to numerous physiological and pathological pathways. HMGB1 is involved in cytokine and chemokine release, leukocyte activation and migration, tumorigenesis, neoangiogenesis, and the activation of several inflammatory pathways. HMGB1 is, in fact, responsible for the trigger, among others, of nuclear factor-κB (NF-κB), tumor necrosis factor-α (TNF-α), toll-like receptor-4 (TLR-4), and vascular endothelial growth factor (VEGF) pathways. Diabetic retinopathy (DR) is a common complication of diabetes mellitus (DM) that is rapidly growing in number. DR is an inflammatory disease caused by hyperglycemia, which determines the accumulation of oxidative stress and cell damage, which ultimately leads to hypoxia and neovascularization. Recent evidence has shown that hyperglycemia is responsible for the hyperexpression of HMGB1. This protein activates numerous pathways that cause the development of DR, and HMGB1 levels are constantly increased in diabetic retinas in both proliferative and non-proliferative stages of the disease. Several molecules, such as glycyrrhizin (GA), have proven effective in reducing diabetic damage to the retina through the inhibition of HMGB1. The main focus of this review is the growing amount of evidence linking HMGB1 and DR as well as the new therapeutic strategies involving this protein.
Collapse
Affiliation(s)
- Marcella Nebbioso
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Alessandro Lambiase
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
- Correspondence: ; Tel.: +39-06-4997-5357; Fax: +39-06-4997-5425
| | - Marta Armentano
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Giosuè Tucciarone
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Vincenza Bonfiglio
- Department of Ophthalmology, University of Catania, Via S. Sofia 76, 95100 Catania, Italy;
| | - Rocco Plateroti
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| | - Ludovico Alisi
- Department of Sense Organs, Faculty of Medicine and Odontology, Policlinico Umberto I, Sapienza University of Rome, p. le A. Moro 5, 00185 Rome, Italy; (M.N.); (M.A.); (G.T.); (R.P.); (L.A.)
| |
Collapse
|
42
|
Mkrtchian S, Kåhlin J, Gómez-Galán M, Ebberyd A, Yoshitake T, Schmidt S, Kehr J, Hildenborg M, Jonsson Fagerlund M, Erlandsson Harris H, Eriksson LI. The impact of damage-associated molecular patterns on the neurotransmitter release and gene expression in the ex vivo rat carotid body. Exp Physiol 2020; 105:1634-1647. [PMID: 32652583 DOI: 10.1113/ep088705] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 07/08/2020] [Indexed: 12/21/2022]
Abstract
NEW FINDINGS What is the central question of this study? Are carotid bodies (CBs) modulated by the damage-associated molecular patterns (DAMPs) and humoral factors of aseptic tissue injury? What are the main findings and their importance? DAMPs (HMGB1, S100 A8/A9) and blood plasma from rats subjected to tibia surgery, a model of aseptic injury, stimulate the release of neurotransmitters (ATP, dopamine) and TNF-α from ex vivo rat CBs. All-thiol HMGB1 mediates upregulation of immune-related biological pathways. These data suggest regulation of CB function by endogenous mediators of innate immunity. ABSTRACT The glomus cells of carotid bodies (CBs) are the primary sensors of arterial partial O2 and CO2 tensions and moreover serve as multimodal receptors responding also to other stimuli, such as pathogen-associated molecular patterns (PAMPs) produced by acute infection. Modulation of CB function by excessive amounts of these immunomodulators is suggested to be associated with a detrimental hyperinflammatory state. We have hypothesized that yet another class of immunomodulators, endogenous danger-associated molecular patterns (DAMPs), released upon aseptic tissue injury and recognized by the same pathogen recognition receptors as PAMPs, might modulate the CB activity in a fashion similar to PAMPs. We have tested this hypothesis by exposing rat CBs to various DAMPs, such as HMGB1 (all-thiol and disulfide forms) and S100 A8/A9 in a series of ex vivo experiments that demonstrated the release of dopamine and ATP, neurotransmitters known to mediate CB homeostatic responses. We observed a similar response after incubating CBs with conditioned blood plasma obtained from the rats subjected to tibia surgery, a model of aseptic injury. In addition, we have investigated global gene expression in the rat CB using an RNA sequencing approach. Differential gene expression analysis showed all-thiol HMGB1-driven upregulation of a number of prominent pro-inflammatory markers including Il1α and Il1β. Interestingly, conditioned plasma had a more profound effect on the CB transcriptome resulting in inhibition rather than activation of the immune-related pathways. These data are the first to suggest potential modulation of CB function by endogenous mediators of innate immunity.
Collapse
Affiliation(s)
- Souren Mkrtchian
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jessica Kåhlin
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marta Gómez-Galán
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anette Ebberyd
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Takashi Yoshitake
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Jan Kehr
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Pronexus Analytical AB, Bromma, Sweden
| | - Malin Hildenborg
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Malin Jonsson Fagerlund
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Helena Erlandsson Harris
- Department of Medicine Solna, Section for Rheumatology, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars I Eriksson
- Department of Physiology and Pharmacology, Section for Anesthesiology and Intensive Care Medicine, Karolinska Institutet, Stockholm, Sweden.,Function Perioperative Medicine and Intensive Care, Karolinska University Hospital and Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
43
|
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
|
44
|
Sun S, Ma J, Xie P, Wu Z, Tian X. Hypoxia-responsive miR-141-3p is involved in the progression of breast cancer via mediating the HMGB1/HIF-1α signaling pathway. J Gene Med 2020; 22:e3230. [PMID: 32436353 PMCID: PMC7685107 DOI: 10.1002/jgm.3230] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022] Open
Abstract
Background Hypoxia‐responsive miRs have been frequently reported in the growth of various malignant tumors. The present study aimed to investigate whether hypoxia‐responsive miR‐141–3p was implicated in the pathogenesis of breast cancer via mediating the high‐mobility group box protein 1 (HMGB1)/hypoxia‐inducible factor (HIF)‐1α signaling pathway. Materials and methods miRs expression profiling was filtrated by miR microarray assays. Gene and protein expression levels, respectively, were examined by a quantitative reverse transcriptase‐polymerase chaion reaction and western blotting. Cell migration and invasion were analyzed using a transwell assay. Cell growth was determined using nude‐mouse transplanted tumor experiments. Results miR‐141–3p was observed as a hypoxia‐responsive miR in breast cancer. miR‐141–3p was down‐regulated in breast cancer specimens and could serve as an independent prognostic factor for predicting overall survival in breast cancer patients. In addition, the overexpression of miR‐141–3p could inhibit hypoxia‐induced cell migration and impede human breast cancer MDA‐MB‐231 cell growth in vivo. Mechanistically, the hypoxia‐related HMGB1/HIF‐1α signaling pathway might be a possible target of miR‐141–3p with respect to preventing the development of breast cancer. Conclusions Our finding provides a new mechanism by which miR‐141–3p could prevent hypoxia‐induced breast tumorigenesis via post‐transcriptional repression of the HMGB1/HIF‐1α signaling pathway.
Collapse
Affiliation(s)
- Shanping Sun
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China.,Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Jinglin Ma
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Panpan Xie
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Zhen Wu
- Department of Breast and Thyroid Surgery, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | - Xingsong Tian
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital, Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
45
|
Yuan X, Bhat OM, Lohner H, Zhang Y, Li PL. Downregulation of Lysosomal Acid Ceramidase Mediates HMGB1-Induced Migration and Proliferation of Mouse Coronary Arterial Myocytes. Front Cell Dev Biol 2020; 8:111. [PMID: 32211403 PMCID: PMC7076051 DOI: 10.3389/fcell.2020.00111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/10/2020] [Indexed: 01/07/2023] Open
Abstract
High-mobility group box 1 protein (HMGB1) has been reported to trigger lysosome destabilization causing a wide of inflammatory diseases. The present study tested whether a lysosomal enzyme, acid ceramidase (AC), plays a critical role in HMGB1-induced alteration in ceramide metabolism and whether such HMGB1-AC interaction is associated with abnormal migration and proliferation of vascular smooth muscle cells (SMCs). We first observed that the expression of AC in the medial layer of mouse coronary arterial wall and colocalization of AC with a lysosome marker Lamp-1. In primary cultured coronary arterial myocytes (CAMs), AC expression and colocalization with Lamp-1 were significantly up-regulated by AC inducer, genistein, but down-regulated by AC inhibitor, N-oleoylethanolamine (NOE). HMGB1 dose-dependently decreased the colocalization of AC with Lamp-1 and reduced mRNA and protein expressions of AC in CAMs, but reversed by genistein. Consistently, HMGB1 significantly induced increases in the levels of long-chain ceramides in CAMs, which were not further enhanced by NOE but blocked by genistein. More importantly, HMGB1 promoted migration and proliferation of CAMs, which were not further increased by NOE but reduced by genistein. Lastly, CAMs isolated from smooth muscle-specific AC knockout mice (AC gene Asah1) exhibited increased ceramide levels and enhanced the migration and proliferation, which resembles the effects of HMGB1 on wild-type CAMs. Together, these results suggest that HMGB1 promotes SMC migration and proliferation via inhibition of AC expression and ceramide accumulation.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Owais M. Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Hannah Lohner
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
46
|
Yao Y, Yao Y, Demetriades AM, Sui A, Su T, Zhu Y, Shen X, Xie B. Neutralization of Bombina variegata peptide 8 suppresses retinal neovascularization in two different murine models: The oxygen-induced retinopathy model and the rhodopsin promoter/VEGF transgenic mouse model. Exp Eye Res 2020; 193:107993. [PMID: 32147400 DOI: 10.1016/j.exer.2020.107993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 02/15/2020] [Accepted: 03/04/2020] [Indexed: 11/25/2022]
Abstract
Bombina variegata 8 (Bv8), also known as prokineticin-2 (PK-2), is a potent pro-angiogenic factor. However, its role in retinal neovascularization (RNV) remains unknown. In this study, we explored the role of Bv8 in the pathogenesis of RNV. We found that the expression of Bv8 was significantly increased in two different models of retinal neovascularization: the oxygen-induced retinopathy (OIR) mouse model and the rhodopsin promoter (rho)/VEGF transgenic mouse model. Neutralization of Bv8 by intravitreal injections of its antibody, not only inhibited retinal and subretinal neovascularization but also decreased the mRNA and protein levels of several pro-angiogenic factors. Our in vitro assay showed that recombinant human Bv8 (RhBv8) protein promoted human retinal microvascular endothelial cells (HRECs) tube-formation, cell proliferation and vascular endothelial growth factor receptor 1 (VEGFR1) and receptor 2 (VEGFR2) expression. Our findings suggest that Bv8 could be used as a novel target for the treatment of RNV-related ocular diseases.
Collapse
Affiliation(s)
- Yiyun Yao
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yixuan Yao
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Anna M Demetriades
- Department of Ophthalmology, New York Presbyterian Hospital-Weill Cornell Medicine, New York, United States
| | - Ailing Sui
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Ting Su
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yanji Zhu
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Xi Shen
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China.
| | - Bing Xie
- Department of Ophthalmology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China.
| |
Collapse
|
47
|
Activation of GPR43 suppresses TNF-α-induced inflammatory response in human fibroblast-like synoviocytes. Arch Biochem Biophys 2020; 684:108297. [PMID: 32035098 DOI: 10.1016/j.abb.2020.108297] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/22/2020] [Accepted: 02/04/2020] [Indexed: 02/07/2023]
Abstract
Although rheumatoid arthritis (RA) has long posed a major threat to global health, the mechanisms driving the development and progression of RA remain incompletely understood. In the present study, we investigated the effects of G protein-coupled receptor 43 (GPR43/FFAR2) in various aspects of the pathogenesis of RA. To our knowledge, this is the first study to demonstrate that GPR43 is expressed on human fibroblast-like synoviocytes (FLS). Furthermore, we show that GPR43 is upregulated in FLS exposed to tumor necrosis factor-α (TNF-α). Importantly, our findings demonstrate that activation of GPR43 using its specific agonist significantly suppressed expression of the following key factors of RA: cytokines, such as interleukin-6 (IL-6), IL-8, high mobility group protein 1 (HMG-1); chemokines, such as monocyte chemoattractant protein 1 (MCP-1), intercellular adhesion molecule 1 (ICAM-1), and vascular cellular adhesion molecule 1 (VCAM-1); markers of oxidative stress, such as production of reactive oxygen species (ROS) and 4-hydroxynoneal (4-HNE); degradative enzymes, such as matrix metalloproteinase-3 (MMP-3) and MMP-13; and activation of the nuclear factor-κB (NF-κB) inflammatory signaling pathway. These results suggest a promising potential role for GPR43 as a specific target in the treatment and prevention of RA.
Collapse
|
48
|
A peptide derived from the core β-sheet region of TIRAP decoys TLR4 and reduces inflammatory and autoimmune symptoms in murine models. EBioMedicine 2020; 52:102645. [PMID: 32014819 PMCID: PMC6997517 DOI: 10.1016/j.ebiom.2020.102645] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 01/08/2020] [Accepted: 01/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND TLRs are some of the actively pursued drug-targets in immune disorders. Owing to a recent surge in the cognizance of TLR structural biology and signalling pathways, numerous therapeutic modulators, ranging from low-molecular-weight organic compounds to polypeptides and nucleic acid agents have been developed. METHODS A penetratin-conjugated small peptide (TIP3), derived from the core β-sheet of TIRAP, was evaluated in vitro by monitoring the TLR-mediated cytokine induction and quantifying the protein expression using western blot. The therapeutic potential of TIP3 was further evaluated in TLR-dependent in vivo disease models. FINDINGS TIP3 blocks the TLR4-mediated cytokine production through both the MyD88- and TRIF-dependent pathways. A similar inhibitory-effect was exhibited for TLR3 but not on other TLRs. A profound therapeutic effect was observed in vivo, where TIP3 successfully alleviated the inflammatory response in mice model of collagen-induced arthritis and ameliorated the disease symptoms in psoriasis and SLE models. INTERPRETATION Our data suggest that TIP3 may be a potential lead candidate for the development of effective therapeutics against TLR-mediated autoimmune disorders. FUNDING This work was supported by the National Research Foundation of Korea (NRF-2019M3A9A8065098, 2019M3D1A1078940 and 2019R1A6A1A11051471). The funders did not have any role in the design of the present study, data collection, data analysis, interpretation, or the writing of the manuscript.
Collapse
|
49
|
Hu Y, Zhang T, Chen J, Cheng W, Chen J, Zheng Z, Lin J, Zhu G, Zhang Y, Bai X, Wang Y, Song B, Wang Q, Qin L, Zhang P. Downregulation of Hypoxia-Inducible Factor-1α by RNA Interference Alleviates the Development of Collagen-Induced Arthritis in Rats. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 19:1330-1342. [PMID: 32160704 PMCID: PMC7038004 DOI: 10.1016/j.omtn.2020.01.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/09/2020] [Accepted: 01/10/2020] [Indexed: 12/13/2022]
Abstract
Rheumatoid arthritis (RA) is the most common type of autoimmune arthritis. Hypoxia-inducible factor-1α (HIF-1α) as a transcription factor in response to hypoxia suggests that it could be a potential therapeutic target for the treatment of RA. In this study, we assessed whether the HIF pathway blockade attenuates the manifestations of RA in the collagen-induced arthritis (CIA) rat model. We constructed a short hairpin RNA (shRNA) lentiviral expression vector targeting HIF-1α (pLVX-shRNA-HIF-1α) and to achieve HIF-1α RNA interference. Quantitative RT-PCR, immunofluorescence staining, and western blot were used to detect the expressions of HIF-1α, vascular endothelial growth factor (VEGF), phsopho (p)-p65, and p-IКBɑ mRNA and protein, respectively. Micro-computed tomography was used to investigate joint morphology at different time points after CIA induction. Moreover, enzyme-linked immunosorbent assay (ELISA) was used to monitor the expression of inflammatory cytokines. In vitro analyses revealed that pLVX-shRNA-HIF-1α effectively inhibited the expression of HIF-1α and VEGF and led to the activation of p-65 and p-IКBɑ, as well as decreased proinflammatory cytokine expression in cell culture. Inhibition of HIF-1α in rats decreased signs of a systemic inflammatory condition, together with decreased pathological changes of RA. Moreover, downregulation of HIF-1α expression markedly reduced the synovitis and angiogenesis. In conclusion, we have shown that pharmacological inhibition of HIF-1 may improve the clinical manifestations of RA.
Collapse
Affiliation(s)
- Yiping Hu
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Tiantian Zhang
- Department of Rheumatology, People's Hospital of Bao'an District, Shenzhen, Guangdong 518128, China
| | - Jingqin Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - WenXiang Cheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Jianhai Chen
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Zhengtan Zheng
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Jietao Lin
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China
| | - Guoyuan Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Yong Zhang
- Shenzhen Pingle Orthopaedic Hospital, Guangdong 518000, China
| | - Xueling Bai
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Yan Wang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China
| | - Bing Song
- School of Dentistry, Cardiff University, Cardiff, Heath Park, CF23 6AL Wales, UK
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China.
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shen Zhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen, Guangdong 518055, China; University of Chinese Academy of Sciences, Beijing 10049, China; University of Chinese Academy of Sciences, Shenzhen Hospital, Shenzhen 518000, China.
| |
Collapse
|
50
|
Shu Z, Miao X, Tang T, Zhan P, Zeng L, Jiang Y. The GSK‑3β/β‑catenin signaling pathway is involved in HMGB1‑induced chondrocyte apoptosis and cartilage matrix degradation. Int J Mol Med 2020; 45:769-778. [PMID: 31922219 PMCID: PMC7015138 DOI: 10.3892/ijmm.2020.4460] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/10/2019] [Indexed: 01/06/2023] Open
Abstract
Knee osteoarthritis (KOA) is a common joint disease with a high incidence rate among middle‑aged and elderly individuals. However, the precise underlying pathological mechanisms and effective treatment of this disease remain to be determined. To explore the effect of high mobility group box 1 (HMGB1) on chondrocyte apoptosis and catabolism, the ATDC5 cell line was cultured as an in vitro model for cartilage research. Cultured cells were treated with recombinant HMGB1 at different concentrations. Hoechst staining and flow cytometry demonstrated that HMGB1 administration significantly induced apoptosis of ATDC5 cells, which was the same as the effect of interleukin‑1β treatment. HMGB1 also induced cartilage matrix degradation, as shown by Alcian blue staining. Moreover, HMGB1 markedly upregulated the expression levels of matrix metallopeptidases (MMPs) and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS), while genetic silencing of HMGB1 significantly suppressed their expressions. The glycogen synthase kinase (GSK)‑3β/β‑catenin pathway was activated upon HMGB1 treatment. Pharmacological inhibitors or HMGB1 knockdown inactivated the GSK‑3β/β‑catenin pathway, inhibited the expression levels of downstream genes, including MMPs and ADAMTS, and attenuated the apoptosis of ATDC5 cells. Furthermore, the data demonstrated that HMGB1 promoted chondrocyte dysfunction via the regulation of estrogen sulfotransferase and Runt‑related transcription factor 2. Thus, the findings of the present study demonstrated that HMGB1 induces chondrocyte cell apoptosis via activation of GSK‑3β/β‑catenin and the subsequent expression of multiple targeted genes.
Collapse
Affiliation(s)
- Zhiyong Shu
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Xiaogang Miao
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Tainhua Tang
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Peng Zhan
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Langqing Zeng
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| | - Yuwen Jiang
- Department of Orthopedics, Zhuhai People's Hospital, Zhuhai, Guangdong 519000, P.R. China
| |
Collapse
|