1
|
Chen W, Wu X, Yaqoob MD, Liu K, Hu Y, Ke X, Hu Y. Analysis of the effect of ALA-PDT on macrophages in footpad model of mice infected with Fonsecaea monophora based on single-cell sequencing. Open Med (Wars) 2025; 20:20241132. [PMID: 39822988 PMCID: PMC11737366 DOI: 10.1515/med-2024-1132] [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: 09/19/2024] [Revised: 12/11/2024] [Accepted: 12/13/2024] [Indexed: 01/19/2025] Open
Abstract
Chromoblastomycosis (CBM) is a chronic neglected fungal disease that causes serious damage to the physical and mental health of patients. 5-Aminolevulinic acid photodynamic therapy (ALA-PDT) has garnered significant attention in the recent era for the treatment of CBM and has exhibited promising effects in several clinical case reports. We established a mice footpad infection model with Fonsecaea monophora and analyzed the impact of PDT treatment on the immune response of macrophages using single-cell sequencing. The results showed that infection of the mouse footpad skin with F. monophora results in an increase in inflammatory cells, primarily mononuclear-macrophages, with the activation of complement and enhancement of cell chemotaxis, leading to upregulation of anti-infection-related pathways. After ALA-PDT treatment, the number of inflammatory cells decreased, while macrophages upregulated the expression of antigen-recognition-related genes, enhancing phagocytosis and autophagy-related biological functions.
Collapse
Affiliation(s)
- Wenyi Chen
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Xuelin Wu
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Muhammad Danish Yaqoob
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Kangxing Liu
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yanqing Hu
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Xiuling Ke
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
| | - Yongxuan Hu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Guangzhou, China
- Department of Dermatology and Venereology, The Third Affiliated Hospital, Southern Medical University, 183 West Zhongshan Road, Guangzhou, China
| |
Collapse
|
2
|
Kara A, Balaban ÖD, Çınar B, Oluğ ST, Kılıç C, Karamustafalıoğlu N. High Mobility Group Box 1 Levels as an Inflammatory Mediator in Bipolar Mania. Noro Psikiyatr Ars 2024; 61:160-166. [PMID: 38868848 PMCID: PMC11165612 DOI: 10.29399/npa.28599] [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: 09/01/2023] [Accepted: 10/12/2023] [Indexed: 06/14/2024] Open
Abstract
Introduction High mobility group box 1 protein (HMGB1) is a member of the molecular family known as damage-associated molecular patterns, which is implicated to have a role in neuroinflammation processes. In recent years, a growing number of studies have focused on the role of inflammation in Bipolar Disorder (BD). This study aimed to investigate the serum levels of HMGB1 and other inflammatory markers in patients with bipolar manic episodes compared to those in healthy controls (HC). Methods A single-center, observational, case-control study was conducted. Thirty-five patients with BD in manic episodes and 35 HC were assessed. Young Mania Rating Scale (YMRS) was used to assess the symptom severity of the patient group. While inflammatory markers (such as HMGB1, C-reactive protein (CRP) and white blood cell count) were assessed at the first three and the last day of hospitalization in the patient group, they were evaluated once in HC. Levels of inflammatory markers were compared between (patient-HC) and within groups (before-after treatment). Results No difference was observed in serum HMGB1 levels of bipolar patients with manic episodes compared to the HC (p>0.05). C-reactive protein levels of manic patients were higher than HC (p<0.001), and the difference persisted even after treatment (p=0.007). In addition, there was a significant positive correlation between CRP levels and antipsychotic drug dosage (r=0.382, p=0.024). Conclusion There were no differences in HMGB1 levels between bipolar patients with acute manic episode and HC. However, higher CRP levels in bipolar patients support the low-grade inflammation hypothesis in the etiology of BD.
Collapse
Affiliation(s)
- Aysu Kara
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| | - Özlem Devrim Balaban
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| | - Bilge Çınar
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| | - Seda Tanrıverdi Oluğ
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| | - Cenk Kılıç
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| | - Nesrin Karamustafalıoğlu
- Bakırköy Prof. Dr. Mazhar Osman Mental Health and Nervous Disorders Training and Research Hospital, Department of Psychiatry, İstanbul, Turkey
| |
Collapse
|
3
|
Liu TT, Sun HF, Han YX, Zhan Y, Jiang JD. The role of inflammation in silicosis. Front Pharmacol 2024; 15:1362509. [PMID: 38515835 PMCID: PMC10955140 DOI: 10.3389/fphar.2024.1362509] [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: 12/28/2023] [Accepted: 02/21/2024] [Indexed: 03/23/2024] Open
Abstract
Silicosis is a chronic illness marked by diffuse fibrosis in lung tissue resulting from continuous exposure to SiO2-rich dust in the workplace. The onset and progression of silicosis is a complicated and poorly understood pathological process involving numerous cells and molecules. However, silicosis poses a severe threat to public health in developing countries, where it is the most prevalent occupational disease. There is convincing evidence supporting that innate and adaptive immune cells, as well as their cytokines, play a significant role in the development of silicosis. In this review, we describe the roles of immune cells and cytokines in silicosis, and summarize current knowledge on several important inflammatory signaling pathways associated with the disease, aiming to provide novel targets and strategies for the treatment of silicosis-related inflammation.
Collapse
Affiliation(s)
| | | | | | - Yun Zhan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | |
Collapse
|
4
|
Lin L, Li J, Song Q, Cheng W, Chen P. The role of HMGB1/RAGE/TLR4 signaling pathways in cigarette smoke-induced inflammation in chronic obstructive pulmonary disease. Immun Inflamm Dis 2022; 10:e711. [PMID: 36301039 PMCID: PMC9552978 DOI: 10.1002/iid3.711] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/08/2022] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a common chronic respiratory disease with irreversible and continuous progression. It has become the fifth most burdensome disease and the third most deadly disease globally. Therefore, the prevention and treatment of COPD are urgent, and it is also important to clarify the pathogenesis of it. Smoking is the main and most common risk factor for COPD. Cigarette smoke (CS) can cause lung inflammation and other pathological mechanisms in the airways and lung tissue. Airway inflammation is one of the important mechanisms leading to the pathogenesis of COPD. Recent studies have shown that high mobility group box 1 (HMGB1) is involved in the occurrence and development of respiratory diseases, including COPD. HMGB1 is a typical damage-associated molecular pattern (DAMP) protein, which mainly exerts its activity by binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4) and further participate in the process of airway inflammation. Studies have shown that the abnormal expression of HMGB1, RAGE, and TLR4 are related to inflammation in COPD. Herein, we discuss the roles of HMGB1, RAGE, and TLR4 in CS/cigarette smoke extract-induced inflammation in COPD, providing a new target for the diagnosis, treatment and prevention of COPD.
Collapse
Affiliation(s)
- Ling Lin
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Jing Li
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Qing Song
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Wei Cheng
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| | - Ping Chen
- Department of Respiratory and Critical Care Medicine, The Second Xiangya HospitalCentral South UniversityChangshaHunanChina
- Research Unit of Respiratory DiseaseCentral South UniversityChangshaHunanChina
- Diagnosis and Treatment Center of Respiratory DiseaseCentral South UniversityChangshaHunanChina
| |
Collapse
|
5
|
Hou W, Zhang H, Jiang M, Wu Y, Li T, Cong L, Duan J. Gu-Ben-Zhi-Ke-Zhong-Yao Alleviated PM2.5-Induced Lung Injury via HMGB1/NF- κB Axis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:8450673. [PMID: 35399858 PMCID: PMC8986406 DOI: 10.1155/2022/8450673] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/08/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022]
Abstract
Background Inhalation of particles with a diameter of less than 2.5 μm (PM2.5) among air pollutants may cause lung damage. Gu-Ben-Zhi-Ke-Zhong-Yao (GBZK) is a traditional Chinese medicine prescription that has a beneficial effect on the treatment of chronic obstructive pulmonary disease (COPD). However, the effect of GBZK on PM2.5-induced lung injury remains to be elucidated. Methods We constructed a mice lung injury model through PM2.5 stimulation and simultaneously performed GBZK gavage treatment. After 4 weeks, the lung tissues of the mice were collected for pathological staining to analyze the degree of damage. The activities of myeloperoxidase (MPO), malondialdehyde (MDA), and oxidative stress-related factors (superoxide dismutase, SOD; glutathione peroxidase, GSH-Px) were detected by commercial kit in lung tissue. Furthermore, the number of neutrophils and related inflammatory factors (interleukin-1, IL-1β; tumor necrosis factor α, TNF-α; interleukin-6, IL-6) in bronchoalveolar lavage fluid (BALF) and serum were collected and tested to evaluate the effect of GBZK on inflammation. Masson staining was used to detect the level of lung fibrosis in mice. The activation of HMGB1 (high-mobility group protein 1) and NFκBp65 (nucleus factor kappa B) in lung tissue was evaluated by immunohistochemistry and western blot. Results The result revealed that PM2.5 induces lung damage, and GBZK gavage treatment could reduce the degree of injury in a concentration-dependent manner in mice. After GBZK treatment, the MPO activity, MDA content, and oxidative stress level in the lung tissues of mice decreased. And after GBZK treatment, the expression levels of inflammatory cytokines in BALF and blood were decreased. GBZK treatment also improved pulmonary fibrosis in mice. In addition, we also found that GBZK prevented the up-regulation of the HMGB1/NF-κB axis in the lungs of mice. Conclusion These results indicated that GBZK might protect mice from PM2.5-induced lung injury by inhibiting the HMGB1/NFκB pathway, thus repressing inflammation and pulmonary fibrosis.
Collapse
Affiliation(s)
- Wenxiao Hou
- Graduate School of Beijing University of Traditional Chinese Medicine, Beijing 100029, China
- Surgical Intensive Care Unit, China Japan Friendship Hospital, Beijing 100029, China
| | - Hongchun Zhang
- Department of Health Care, China Japan Friendship Hospital, Beijing 100029, China
- National Respiratory Center, Beijing 100029, China
| | - Meng Jiang
- Yunnan University of Traditional Chinese Medicine, Kunming 650011, China
| | - Yina Wu
- Surgical Intensive Care Unit, China Japan Friendship Hospital, Beijing 100029, China
| | - Tao Li
- Surgical Intensive Care Unit, China Japan Friendship Hospital, Beijing 100029, China
| | - Luhong Cong
- Surgical Intensive Care Unit, China Japan Friendship Hospital, Beijing 100029, China
- Department of Emergency, China Japan Friendship Hospital, Beijing 100029, China
| | - Jun Duan
- Surgical Intensive Care Unit, China Japan Friendship Hospital, Beijing 100029, China
- Department of Emergency, China Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
6
|
Marti JLG, Wells A, Brufsky AM. Dysregulation of the mevalonate pathway during SARS-CoV-2 infection: An in silico study. J Med Virol 2021; 93:2396-2405. [PMID: 33331649 PMCID: PMC9553089 DOI: 10.1002/jmv.26743] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022]
Abstract
SARS-CoV-2 triggers a dysregulated innate immune system activation. As the mevalonate pathway (MVP) prevents the activation of inflammasomes and cytokine release and regulates endosomal transport, compromised signaling could be associated with the pathobiology of COVID-19. Prior transcriptomic studies of host cells in response to SARS-CoV-2 infection have not reported to date the effects of SARS-CoV-2 on the MVP. In this study, we accessed public data sets to report in silico investigations into gene expression. In addition, we proposed candidate genes that are thought to have a direct association with the pathogenesis of COVID-19, and which may be dependent on signals derived from the MVP. Our results revealed dysregulation of genes involved in the MVP. These results were not found when investigating the gene expression data from host cells infected with H3N2 influenza virus, H1N1 influenza virus, or respiratory syncytial virus. Our manually curated gene set showed significant gene expression variability in A549 cells infected with SARS-CoV-2, as per Blanco-Melo et al. data set (GSE147507). In light of the present findings, SARS-CoV-2 could hijack the MVP, leading to hyperinflammatory responses. Prompt reconstitution of this pathway with available agents should be considered in future studies.
Collapse
Affiliation(s)
- Juan Luis Gomez Marti
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Health System, Pittsburgh, Pennsylvania, USA
| | - Alan Wells
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Pittsburgh VA Health System, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Adam M. Brufsky
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
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
|
8
|
Yu Y, Ou-Yang W, Zhang H, Jiang T, Cho WC, Zhu H, Xiao Z, Li S. High-mobility Group Box 1 Facilitates CD4 T Cell Self-aggregation Via Integrin and STAT3 Activation Before Homing. Inflamm Bowel Dis 2020; 26:1188-1198. [PMID: 32130404 DOI: 10.1093/ibd/izaa044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) is one of the delayed pro-inflammatory cytokines produced in the later stages of pathogenesis and plays an important role in the progression of various inflammatory and autoimmune diseases. High-mobility group box 1 is able to stimulate interaction between integrins and cell adhesion molecules to facilitate cell-cell aggregation in "tissue-specific" endothelium; however, whether and how HMGB1 affects the adhesive capability of early acting immune cells in bloodstream remains largely unknown. METHODS Human peripheral blood samples were collected from healthy adult donors. The CD4 T cells were isolated from blood using CD4 T cell isolation kit and identified using flow cytometry and immunofluorescence staining. The effect of HMGB1 on adhesive ability of CD4 T cells was accessed by cell self-aggregation assay and endothelial adhesion assay. The migratory ability of CD4 T cells was evaluated by cell migration assay. Secretion of pro-inflammatory cytokines or chemokine C-X-C motif chemokine 12 (CXCL12) were detected by ELISA. Expression of integrins β1, β7, and α4β7 were determined by flow cytometric analysis. Inhibition of integrins was achieved with anti-integrin antibodies or cyclic peptide inhibitors. Activation of signal transducers and activators of transcription 3 (STAT3) was measured by flow cytometry and fluorescent staining. RESULTS High-mobility group box 1 facilitated CD4 T cell self-aggregation with simultaneous reduction of CD4 T single-cell counts in the bloodstream. The CD4 T cell self-aggregation induced by HMGB1 resulted in upregulation of integrins β1, β7, and α4β7; release of other pro-inflammatory cytokines or chemokine CXCL12; and activation of STAT3 signaling. Intriguingly, pro-inflammatory cytokines induced by HMGB1 could further amplify CD4 T cell self-aggregation. HMGB1 induced CD4 T cell apoptosis via activation of caspase-3/7. Furthermore, HMGB1 promoted migration and adhesion of CD4 T cells to endothelial cells. CONCLUSIONS These results provide proof of concept that HMGB1 promotes CD4 T cell self-aggregation before homing to inflammatory sites and highlight the potential of blocking immune cell self-aggregation in blood as a novel therapeutic approach against the development and progression of HMGB1-related inflammatory diseases.HMGB1 induces CD4 T cell self-aggregation in blood resulting in upregulation of integrins expression and release of pro-inflammatory cytokines/chemokines via activation of STAT3 signaling. This study highlights the potential of preventive and therapeutic intervention on immune cell self-aggregation in the bloodstream.
Collapse
Affiliation(s)
- Ying Yu
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - Wenxian Ou-Yang
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - Hui Zhang
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - Tao Jiang
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong
| | - Huang Zhu
- Department of Medicine, Division of Regenerative Medicine, University of California-San Diego, San Diego, California, USA
| | - Zhenghui Xiao
- Emergency Center of Hunan Children's Hospital, Changsha, China
| | - Shuangjie Li
- Department of Hepatopathy, Hunan Children's Hospital, Changsha, China
| |
Collapse
|
9
|
Koshkin V, Bleker de Oliveira M, Peng C, Ailles LE, Liu G, Covens A, Krylov SN. Spheroid-Based Approach to Assess the Tissue Relevance of Analysis of Dispersed-Settled Tissue Cells by Cytometry of the Reaction Rate Constant. Anal Chem 2020; 92:9348-9355. [PMID: 32522000 DOI: 10.1021/acs.analchem.0c01667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytometry of Reaction Rate Constant (CRRC) uses time-lapse fluorescence microscopy to measure a rate constant of a catalytic reaction in individual cells and, thus, facilitate accurate size determination for cell subpopulations with distinct efficiencies of this reaction. Reliable CRRC requires uniform exposure of cells to the reaction substrate followed by their uniform imaging, which in turn, requires that a tissue sample be disintegrated into a suspension of dispersed cells, and these cells settle on the support surface before being analyzed by CRRC. We call such cells "dispersed-settled" to distinguish them from cells cultured as a monolayer. Studies of the dispersed-settled cells can be tissue-relevant only if the cells maintain their 3D tissue state during the multi-hour CRRC procedure. Here, we propose an approach for assessing tissue relevance of the CRRC-based analysis of the dispersed-settled cells. Our approach utilizes cultured multicellular spheroids as a 3D cell model and cultured cell monolayers as a 2D cell model. The CRRC results of the dispersed-settled cells derived from spheroids are compared to those of spheroids and monolayers in order to find if the dispersed-settled cells are representative of the spheroids. To demonstrate its practical use, we applied this approach to a cellular reaction of multidrug resistance (MDR) transport, which was followed by extrusion of a fluorescent substrate from the cells. The approach proved to be reliable and revealed long-term maintenance of MDR transport in the dispersed-settled cells obtained from cultured ovarian cancer spheroids. Accordingly, CRRC can be used to determine accurately the size of a cell subpopulation with an elevated level of MDR transport in tumor samples, which makes CRRC a suitable method for the development of MDR-based predictors of chemoresistance. The proposed spheroid-based approach for validation of CRRC is applicable to other types of cellular reactions and, thus, will be an indispensable tool for transforming CRRC from an experimental technique into a practical analytical tool.
Collapse
Affiliation(s)
- Vasilij Koshkin
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | | | - Chun Peng
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Laurie E Ailles
- Princess Margaret Cancer Centre and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario N5G 1L7, Canada
| | - Geoffrey Liu
- Department of Medicine, Medical Oncology, Princess Margaret Cancer Centre, Toronto, Ontario M5G 2M9, Canada
| | - Allan Covens
- Sunnybrook Odette Cancer Centre, Toronto, Ontario M4N 3M5, Canada
| | - Sergey N Krylov
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
10
|
Khambu B, Yan S, Huda N, Yin XM. Role of High-Mobility Group Box-1 in Liver Pathogenesis. Int J Mol Sci 2019; 20:ijms20215314. [PMID: 31731454 PMCID: PMC6862281 DOI: 10.3390/ijms20215314] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 10/16/2019] [Accepted: 10/23/2019] [Indexed: 12/21/2022] Open
Abstract
High-mobility group box 1 (HMGB1) is a highly abundant DNA-binding protein that can relocate to the cytosol or undergo extracellular release during cellular stress or death. HMGB1 has a functional versatility depending on its cellular location. While intracellular HMGB1 is important for DNA structure maintenance, gene expression, and autophagy induction, extracellular HMGB1 acts as a damage-associated molecular pattern (DAMP) molecule to alert the host of damage by triggering immune responses. The biological function of HMGB1 is mediated by multiple receptors, including the receptor for advanced glycation end products (RAGE) and Toll-like receptors (TLRs), which are expressed in different hepatic cells. Activation of HMGB1 and downstream signaling pathways are contributing factors in the pathogenesis of non-alcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), and drug-induced liver injury (DILI), each of which involves sterile inflammation, liver fibrosis, ductular reaction, and hepatic tumorigenesis. In this review, we will discuss the critical role of HMGB1 in these pathogenic contexts and propose HMGB1 as a bona fide and targetable DAMP in the setting of common liver diseases.
Collapse
Affiliation(s)
- Bilon Khambu
- Correspondence: ; Tel.: +1-317-274-1789; Fax: +1-317-491-6639
| | | | | | | |
Collapse
|
11
|
Rouhiainen A, Nykänen NP, Kuja-Panula J, Vanttola P, Huttunen HJ, Rauvala H. Inhibition of Homophilic Interactions and Ligand Binding of the Receptor for Advanced Glycation End Products by Heparin and Heparin-Related Carbohydrate Structures. MEDICINES (BASEL, SWITZERLAND) 2018; 5:E79. [PMID: 30061484 PMCID: PMC6165534 DOI: 10.3390/medicines5030079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/08/2018] [Accepted: 07/23/2018] [Indexed: 01/12/2023]
Abstract
Background: Heparin and heparin-related sulphated carbohydrates inhibit ligand binding of the receptor for advanced glycation end products (RAGE). Here, we have studied the ability of heparin to inhibit homophilic interactions of RAGE in living cells and studied how heparin related structures interfere with RAGE⁻ligand interactions. Methods: Homophilic interactions of RAGE were studied with bead aggregation and living cell protein-fragment complementation assays. Ligand binding was analyzed with microwell binding and chromatographic assays. Cell surface advanced glycation end product binding to RAGE was studied using PC3 cell adhesion assay. Results: Homophilic binding of RAGE was mediated by V₁- and modulated by C₂-domain in bead aggregation assay. Dimerisation of RAGE on the living cell surface was inhibited by heparin. Sulphated K5 carbohydrate fragments inhibited RAGE binding to amyloid β-peptide and HMGB1. The inhibition was dependent on the level of sulfation and the length of the carbohydrate backbone. α-d-Glucopyranosiduronic acid (glycyrrhizin) inhibited RAGE binding to advanced glycation end products in PC3 cell adhesion and protein binding assays. Further, glycyrrhizin inhibited HMGB1 and HMGB1 A-box binding to heparin. Conclusions: Our results show that K5 polysaccharides and glycyrrhizin are promising candidates for RAGE targeting drug development.
Collapse
Affiliation(s)
- Ari Rouhiainen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
- Department of Biosciences, University of Helsinki, 00014 Helsinki, Finland.
| | - Niko-Petteri Nykänen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
- Department of Translational Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), D-81377 Munich, Germany.
| | - Juha Kuja-Panula
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
| | - Päivi Vanttola
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
- Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland.
| | - Henri J Huttunen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
| | - Heikki Rauvala
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland.
| |
Collapse
|
12
|
Wu XJ, Chen YY, Gong CC, Pei DS. The role of high-mobility group protein box 1 in lung cancer. J Cell Biochem 2018; 119:6354-6365. [PMID: 29665052 DOI: 10.1002/jcb.26837] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/09/2018] [Indexed: 12/14/2022]
Abstract
High-mobility group protein box 1(HMGB1)is a ubiquitous highly conserved nuclear protein. Acting as a chromatin-binding factor, HMGB1 binds to DNA and plays an important role in stabilizing nucleosome formation, facilitating gene transcription, DNA repairing, inflammation, cell differentiation, and regulating the activity of steroid hormone receptors. Currently, HMGB1 is discovered to be related to development, progression, and targeted therapy of lung cancer, which makes it an attractive biomarker, and therapeutic target. This review aims to encapsulate the relationship between HMGB1 and lung cancer, suggesting that HMGB1 plays a pivotal role in initiation, development, invasion, metastasis, and prognosis of lung cancer.
Collapse
Affiliation(s)
- Xiao-Jin Wu
- Department of Radiation Oncology, The First People's Hospital of Xuzhou, Xuzhou, China.,Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Yuan-Yuan Chen
- Department of Radiation Oncology, The First People's Hospital of Xuzhou, Xuzhou, China
| | - Chan-Chan Gong
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| | - Dong-Sheng Pei
- Department of Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
13
|
Irradiation Enhances Abscopal Anti-tumor Effects of Antigen-Specific Immunotherapy through Regulating Tumor Microenvironment. Mol Ther 2017; 26:404-419. [PMID: 29248428 DOI: 10.1016/j.ymthe.2017.11.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 11/14/2017] [Accepted: 11/14/2017] [Indexed: 01/23/2023] Open
Abstract
Ionizing radiation therapy is a well-established method of eradicating locally advanced tumors. Here, we examined whether local RT enhanced the potency of an antigen-specific DNA vaccine, and we investigated the possible underlying mechanism. Using the HPV16 E6/E7+ syngeneic TC-1 tumor, we evaluated the combination of CTGF/E7 vaccination with local irradiation with regard to synergistic antigen-specific immunity and anti-tumor effects. Tumor-bearing mice treated with local RT (6 Gy twice weekly) and CTGF/E7 DNA vaccination exhibited dramatically increased numbers of E7-specific CD8+ cytotoxic T cell precursors, higher titers of anti-E7 Abs, and significantly reduced tumor size. The combination of local RT and CTGF/E7 vaccination also elicited abscopal effects on non-irradiated local subcutaneous and distant pulmonary metastatic tumors. Local irradiation induced the expression of high-mobility group box 1 protein (HMGB-1) in apoptotic tumor cells and stimulated dendritic cell (DC) maturation, consequently inducing antigen-specific immune responses. Additionally, local irradiation eventually increased the effector-to-suppressor cell ratio in the tumor microenvironment. Overall, local irradiation enhanced the antigen-specific immunity and anti-tumor effects on local and distant metastatic tumors generated by an antigen-specific DNA vaccine. These findings suggest that the combination of irradiation with antigen-specific immunotherapy is a promising new clinical strategy for cancer therapy.
Collapse
|
14
|
Sheller-Miller S, Urrabaz-Garza R, Saade G, Menon R. Damage-Associated molecular pattern markers HMGB1 and cell-Free fetal telomere fragments in oxidative-Stressed amnion epithelial cell-Derived exosomes. J Reprod Immunol 2017; 123:3-11. [PMID: 28858636 DOI: 10.1016/j.jri.2017.08.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/13/2017] [Accepted: 08/09/2017] [Indexed: 01/13/2023]
Abstract
Term labor in humans is associated with increased oxidative stress (OS) -induced senescence and damages to amnion epithelial cells (AECs). Senescent fetal cells release alarmin high-mobility group box 1 (HMGB1) and cell-free fetal telomere fragments (cffTF) which can be carried by exosomes to other uterine tissues to produce parturition-associated inflammatory changes. This study characterized AEC-derived exosomes under normal and OS conditions and their packaging of HMGB1 and cffTF. Primary AECs were treated with either standard media or oxidative stress-induced media (exposure to cigarette smoke extract for 48h). Senescence was determined, and exosomes were isolated and characterized. To colocalize HMGB1 and cffTF in amnion exosomes, immunofluorescent staining and in situ hybridization were performed, followed by confocal microscopy. Next generation sequencing (NGS) determined exosomal cffTF and other cell-free amnion cell DNA specificity. Regardless of condition, primary AECs produce exosomes with a classic size, shape, and markers. OS and senescence caused the translocation of HMGB1 and cffTF from AECs' nuclei to cytoplasm compared to untreated cells, which was inhibited by antioxidant N-acetyl cysteine (NAC). Linescans confirmed colocalization of HMGB1 and cffTF in exosomes were higher in the cytoplasm after CSE treatment compared to untreated AECs. NGS determined that besides cffTF, AEC exosomes also carry genomic and mitochondrial DNA, regardless of growth conditions. Sterile inflammatory markers HMGB1 and cffTF from senescent fetal cells are packaged inside exosomes. We postulate that this exosomal cargo can act as a fetal signal at term and can cause labor-associated changes in neighboring tissues.
Collapse
Affiliation(s)
- Samantha Sheller-Miller
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA; Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Rheanna Urrabaz-Garza
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| | - George Saade
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
15
|
He SJ, Cheng J, Feng X, Yu Y, Tian L, Huang Q. The dual role and therapeutic potential of high-mobility group box 1 in cancer. Oncotarget 2017; 8:64534-64550. [PMID: 28969092 PMCID: PMC5610024 DOI: 10.18632/oncotarget.17885] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 04/24/2017] [Indexed: 12/31/2022] Open
Abstract
High-mobility group box 1 (HMGB1) is an abundant protein in most eukaryocytes. It can bind to several receptors such as advanced glycation end products (RAGE) and Toll-like receptors (TLRs), in direct or indirect way. The biological effects of HMGB1 depend on its expression and subcellular location. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription, telomere maintenance, and genome stability. While outside the nucleus, it possesses more complicated functions, including regulating cell proliferation, autophagy, inflammation and immunity. During tumor development, HMGB1 has been characterized as both a pro- and anti-tumoral protein by either promoting or suppressing tumor growth, proliferation, angiogenesis, invasion and metastasis. However, the current knowledge concerning the positive and negative effects of HMGB1 on tumor development is not explicit. Here, we evaluate the role of HMGB1 in tumor development and attempt to reconcile the dual effects of HMGB1 in carcinogenesis. Furthermore, we would like to present current strategies targeting against HMGB1, its receptor or release, which have shown potentially therapeutic value in cancer intervention.
Collapse
Affiliation(s)
- Si-Jia He
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Cheng
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiao Feng
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Yu
- Oncology Department, Henan Provincial People's Hospital, Zhengzhou, China
| | - Ling Tian
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Huang
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pancreatic Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Appavoo E, Hajam IA, Muneeswaran NS, Kondabattula G, Bhanuprakash V, Kishore S. Synergistic effect of high-mobility group box-1 and lipopolysaccharide on cytokine induction in bovine peripheral blood mononuclear cells. Microbiol Immunol 2017; 60:196-202. [PMID: 26639899 DOI: 10.1111/1348-0421.12350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/14/2015] [Accepted: 12/02/2015] [Indexed: 01/06/2023]
Abstract
High-mobility group box 1 (HMGB1) is one of the potent endogenous adjuvants released by necrotic and activated innate immune cells. HMGB1 modulates innate and adaptive immune responses in humans and mice by mediating immune cells crosstalk. However, the immuno-modulatory effects of HMGB1 in the bovine immune system are not clearly known. In this study, the effect of bovine HMGB1 alone or in combination with LPS on the expression kinetics of cytokines upon in vitro stimulation of bovine peripheral blood mononuclear cells (PBMCs) was investigated by quantitative PCR assay. The biological activity of bovine HMGB1 expressed in this prokaryotic expression system was confirmed by its ability to induce nitric oxide secretion in RAW 264.7 cells. The present results indicate that HMGB1 induces a more delayed TNF-α response than does LPS in stimulated PBMCs. However, IFN-γ, IFN-β and IL-12 mRNA transcription peaked at 6 hr post stimulation after both treatments. Further, HMGB1 and LPS heterocomplex up-regulated TNF-α, IFN-γ and IL-12 mRNA expression significantly than did individual TLR4 agonists. The heterocomplex also enhanced the expression of TLR4 on bovine PBMCs. In conclusion, the data indicate that HMGB1 and LPS act synergistically and enhance proinflammatory cytokines, thereby eliciting Th1 responses in bovine PBMCs. These results suggest that HMGB1 can act as an adjuvant in modulating the bovine immune system and thus lays a foundation for using HMGB1 as an adjuvant in various bovine vaccine preparations.
Collapse
Affiliation(s)
- Elamurugan Appavoo
- Foot and Mouth Disease Research Center, Indian Veterinary Research Institute, Bangalore 560024, India
| | - Irshad Ahmed Hajam
- Foot and Mouth Disease Research Center, Indian Veterinary Research Institute, Bangalore 560024, India
| | | | - Ganesh Kondabattula
- Foot and Mouth Disease Research Center, Indian Veterinary Research Institute, Bangalore 560024, India
| | | | - Subodh Kishore
- Foot and Mouth Disease Research Center, Indian Veterinary Research Institute, Bangalore 560024, India
| |
Collapse
|
17
|
Vogel S, Rath D, Borst O, Mack A, Loughran P, Lotze MT, Neal MD, Billiar TR, Gawaz M. Platelet-derived high-mobility group box 1 promotes recruitment and suppresses apoptosis of monocytes. Biochem Biophys Res Commun 2016; 478:143-148. [PMID: 27449608 DOI: 10.1016/j.bbrc.2016.07.078] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 07/19/2016] [Indexed: 01/22/2023]
Abstract
Platelets are circulating cellular sensors that express and release the damage-associated molecular pattern molecule (DAMP) high-mobility group box 1 (HMGB1) at sites of disrupted vascular and tissue integrity. We have recently identified platelet-derived HMGB1 as a critical mediator of thrombosis. The role of platelet-derived HMGB1 in mediating interactions with monocytes remains unknown. In transgenic mice with platelet-specific ablation of HMGB1 and neutralization studies, we show that HMGB1 derived from platelets promotes recruitment of monocytes and prevents monocytes from undergoing apoptosis. During experimental trauma and hemorrhagic shock, infiltrated monocytes in the lung and liver were significantly attenuated in mice lacking HMGB1 in platelets. Platelet-derived HMGB1 mediated monocyte migration via the receptor for advanced glycation end products (RAGE) and suppressed apoptosis via toll-like receptor 4 (TLR4)-dependent activation of MAPK/ERK (extracellular signal-regulated kinase) in monocytes. In conclusion, we identify platelet-derived HMGB1 as a critical regulator of monocyte recruitment and apoptosis, with potential implications in disease states associated with thrombosis and inflammation.
Collapse
Affiliation(s)
- Sebastian Vogel
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University Tübingen, Germany.
| | - Dominik Rath
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University Tübingen, Germany
| | - Oliver Borst
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University Tübingen, Germany
| | - Andreas Mack
- Institute of Anatomy, Eberhard Karls University Tübingen, Germany
| | - Patricia Loughran
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Matthew D Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Meinrad Gawaz
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University Tübingen, Germany
| |
Collapse
|
18
|
Zeng JC, Xiang WY, Lin DZ, Zhang JA, Liu GB, Kong B, Gao YC, Lu YB, Wu XJ, Yi LL, Zhong JX, Xu JF. Elevated HMGB1-related interleukin-6 is associated with dynamic responses of monocytes in patients with active pulmonary tuberculosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:1341-1353. [PMID: 25973018 PMCID: PMC4396284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 01/23/2015] [Indexed: 06/04/2023]
Abstract
There were limited studies assessing the role of HMGB1 in TB infection. In this prospective study, we aimed to assess the levels of HMGB1 in plasma or sputum from active pulmonary tuberculosis (APTB) patients positive for Mtb culture test, and to evaluate its relationship with inflammatory cytokines and innate immune cells. A total of 36 sputum Mtb culture positive APTB patients and 32 healthy volunteers (HV) were included. Differentiated THP-1 cells were treated for 6, 12 and 24 hrs with BCG at a multiplicity of infection of 10. The absolute values and percentages of white blood cells (WBC), neutrophils, lymphocytes, and monocytes were detected by an automatic blood analyzer. Levels of HMGB1, IL-6, IL-10 and TNF-α in plasma, sputum, or cell culture supernatant were measured by ELISA. The blood levels of HMGB1, IL-6, IL-10 and TNF-α, the absolute values of WBC, monocytes and neutrophils, and the percentage of monocytes were significant higher in APTB patients than those in HV groups (P < 0.05). The sputum levels of HMGB1, IL-10, and TNF-α were also significantly higher in APTB patients than those in HV groups (P < 0.05). Meanwhile, plasma level of HMGB1, IL-6, and IL-10 in APTB patients were positively correlated with those in sputum (P < 0.05), respectively. IL-6 was positively correlated with HMGB1 both in plasma and sputum of APTB patients (P < 0.05). HMGB1 and IL-6 is positively correlated with the absolute number of monocytes in APTB patients (P < 0.05). BCG induced HMGB1, IL-6, IL-10 and TNF-α production effectively in PMA-treated THP-1 cells. HMGB1 may be used as an attractive biomarker for APTB diagnosis and prognosis and may reflect the inflammatory status of monocytes in patients with APTB.
Collapse
Affiliation(s)
- Jin-Cheng Zeng
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics1 Xincheng Road, Dongguan 523808, People’s Republic of China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Wen-Yu Xiang
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Dong-Zi Lin
- Dongguan 6 People’s HospitalDongguan 523008, People’s Republic of China
| | - Jun-Ai Zhang
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Gan-Bin Liu
- Dongguan 6 People’s HospitalDongguan 523008, People’s Republic of China
| | - Bin Kong
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Yu-Chi Gao
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Yuan-Bin Lu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| | - Xian-Jing Wu
- Department of Clinical Laboratory, Affiliated Hospital of Guangdong Medical CollegeZhanjiang 524001, People’s Republic of China
| | - Lai-Long Yi
- Dongguan 6 People’s HospitalDongguan 523008, People’s Republic of China
| | - Ji-Xin Zhong
- Division of Cardiovascular Medicine, University of Maryland School of MedicineMaryland 21201, USA
| | - Jun-Fa Xu
- Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics1 Xincheng Road, Dongguan 523808, People’s Republic of China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical CollegeNo. 1 Xincheng Road, Dongguan 523808, People’s Republic of China
| |
Collapse
|
19
|
Vogel S, Börger V, Peters C, Förster M, Liebfried P, Metzger K, Meisel R, Däubener W, Trapp T, Fischer JC, Gawaz M, Sorg RV. Necrotic cell-derived high mobility group box 1 attracts antigen-presenting cells but inhibits hepatocyte growth factor-mediated tropism of mesenchymal stem cells for apoptotic cell death. Cell Death Differ 2015; 22:1219-30. [PMID: 25571972 DOI: 10.1038/cdd.2014.225] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 11/09/2022] Open
Abstract
Tissue damage due to apoptotic or necrotic cell death typically initiates distinct cellular responses, leading either directly to tissue repair and regeneration or to immunological processes first, to clear the site, for example, of potentially damage-inducing agents. Mesenchymal stem cells (MSC) as well as immature dendritic cells (iDC) and monocytes migrate to injured tissues. MSC have regenerative capacity, whereas monocytes and iDC have a critical role in inflammation and induction of immune responses, including autoimmunity after tissue damage. Here, we investigated the influence of apoptotic and necrotic cell death on recruitment of MSC, monocytes and iDC, and identified hepatocyte growth factor (HGF) and the alarmin high mobility group box 1 (HMGB1) as key factors differentially regulating these migratory responses. MSC, but not monocytes or iDC, were attracted by apoptotic cardiomyocytic and neuronal cells, whereas necrosis induced migration of monocytes and iDC, but not of MSC. Only apoptotic cell death resulted in HGF production and HGF-mediated migration of MSC towards the apoptotic targets. In contrast, HMGB1 was predominantly released by the necrotic cells and mediated recruitment of monocytes and iDC via the receptor of advanced glycation end products. Moreover, necrotic cardiomyocytic and neuronal cells caused an HMGB1/toll-like receptor-4-dependent inhibition of MSC migration towards apoptosis or HGF, while recruitment of monocytes and iDC by necrosis or HMGB1 was not affected by apoptotic cells or HGF. Thus, the type of cell death differentially regulates recruitment of either MSC or monocytes and iDC through HGF and HMGB1, respectively, with a dominant, HMGB1-mediated role of necrosis in determining tropism after tissue injury.
Collapse
Affiliation(s)
- S Vogel
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - V Börger
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - C Peters
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - M Förster
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - P Liebfried
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - K Metzger
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| | - R Meisel
- Clinic of Pediatric Oncology, Hematology and Clinical Immunology, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - W Däubener
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - T Trapp
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - J C Fischer
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - M Gawaz
- Department of Cardiology and Cardiovascular Diseases, Eberhard Karls University, Tübingen, Germany
| | - R V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University Hospital, Düsseldorf, Germany
| |
Collapse
|
20
|
HMGB1 promotes a p38MAPK associated non-infectious inflammatory response pathway in human fetal membranes. PLoS One 2014; 9:e113799. [PMID: 25469638 PMCID: PMC4254744 DOI: 10.1371/journal.pone.0113799] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 10/30/2014] [Indexed: 11/24/2022] Open
Abstract
Objective Spontaneous preterm birth (PTB) and preterm prelabor rupture of membranes (pPROM) are major pregnancy complications often associated with a fetal inflammatory response. Biomolecular markers of this fetal inflammatory response to both infectious and non-infectious risk factors and their contribution to PTB and pPROM mechanism are still unclear. This study examined fetal membrane production, activation and mechanistic properties of high mobility group box 1 (HMGB1) as a contributor of the non-infectious fetal inflammatory response. Materials and Methods HMGB1 transcripts and active HMGB1 were profiled in fetal membranes and amniotic fluids collected from PTB and normal term birth. In vitro, normal term not in labor fetal membranes were exposed to lipopolysaccharide (LPS) and water soluble cigarette smoke extract (CSE). HMGB1-transcripts and its protein concentrations were documented by RT-PCR and ELISA. Recombinant HMGB1 treated membranes and media were subjected to RT-PCR for HMGB1 receptors, mitogen activated protein kinase pathway analysis, cytokine levels, and Western blot for p38MAPK. Results HMGB1 expression and its active forms were higher in PTB and pPROM than normal term membranes and amniotic fluid samples. Both LPS and CSE enhanced HMGB1 expression and release in vitro. Fetal membrane exposure to HMGB1 resulted in increased expression of TLR2 and 4 and dose-dependent activation of p38MAPK-mediated inflammation. Conclusions HMGB1 increase by fetal membrane cells in response to either oxidative stress or infection can provide a positive feedback loop generating non-infectious inflammatory activation. Activation of p38MAPK by HMGB1 promotes development of the senescence phenotype and senescence associated sterile inflammation. HMGB1 activity is an important regulator of the fetal inflammatory response regardless of infection.
Collapse
|
21
|
Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, Huang J, Yu Y, Fan XG, Yan Z, Sun X, Wang H, Wang Q, Tsung A, Billiar TR, Zeh HJ, Lotze MT, Tang D. HMGB1 in health and disease. Mol Aspects Med 2014; 40:1-116. [PMID: 25010388 PMCID: PMC4254084 DOI: 10.1016/j.mam.2014.05.001] [Citation(s) in RCA: 740] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/05/2014] [Indexed: 12/22/2022]
Abstract
Complex genetic and physiological variations as well as environmental factors that drive emergence of chromosomal instability, development of unscheduled cell death, skewed differentiation, and altered metabolism are central to the pathogenesis of human diseases and disorders. Understanding the molecular bases for these processes is important for the development of new diagnostic biomarkers, and for identifying new therapeutic targets. In 1973, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and termed high-mobility group (HMG) proteins. The HMG proteins include three superfamilies termed HMGB, HMGN, and HMGA. High-mobility group box 1 (HMGB1), the most abundant and well-studied HMG protein, senses and coordinates the cellular stress response and plays a critical role not only inside of the cell as a DNA chaperone, chromosome guardian, autophagy sustainer, and protector from apoptotic cell death, but also outside the cell as the prototypic damage associated molecular pattern molecule (DAMP). This DAMP, in conjunction with other factors, thus has cytokine, chemokine, and growth factor activity, orchestrating the inflammatory and immune response. All of these characteristics make HMGB1 a critical molecular target in multiple human diseases including infectious diseases, ischemia, immune disorders, neurodegenerative diseases, metabolic disorders, and cancer. Indeed, a number of emergent strategies have been used to inhibit HMGB1 expression, release, and activity in vitro and in vivo. These include antibodies, peptide inhibitors, RNAi, anti-coagulants, endogenous hormones, various chemical compounds, HMGB1-receptor and signaling pathway inhibition, artificial DNAs, physical strategies including vagus nerve stimulation and other surgical approaches. Future work further investigating the details of HMGB1 localization, structure, post-translational modification, and identification of additional partners will undoubtedly uncover additional secrets regarding HMGB1's multiple functions.
Collapse
Affiliation(s)
- Rui Kang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| | - Ruochan Chen
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Qiuhong Zhang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Wen Hou
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Sha Wu
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Lizhi Cao
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Jin Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yan Yu
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Xue-Gong Fan
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Zhengwen Yan
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA; Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong 510120, China
| | - Xiaofang Sun
- Key Laboratory for Major Obstetric Diseases of Guangdong Province, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Experimental Department of Institute of Gynecology and Obstetrics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Haichao Wang
- Laboratory of Emergency Medicine, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Qingde Wang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Allan Tsung
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Herbert J Zeh
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Michael T Lotze
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA
| | - Daolin Tang
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, USA.
| |
Collapse
|
22
|
Pei JX, Xiong GL, Xie Y. Relationship between high mobility group box 1 protein and inflammatory bowel disease. Shijie Huaren Xiaohua Zazhi 2014; 22:5092-5099. [DOI: 10.11569/wcjd.v22.i33.5092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
High mobility group box 1 protein (HMGB1) is a DNA binding protein that can promote the maintenance of nucleosomal structures and regulate gene transcription in mammalian cells. HMGB1 is a ubiquitous nuclear protein that is widely distributed among mammalian cells, passively released from necrotic cells and actively released from stimulated inflammatory cells. HMGB1 might function as an endogenous immune adjuvant and play a crucial role in the development of various inflammatory diseases, and blockade of HMGB1 expression attenuates the intestinal inflammation and damage in animal models. This article reviews recent progress in understanding the relationship between HMGB1 and inflammatory bowel disease.
Collapse
|
23
|
High-mobility group box 1 (HMGB1) in childhood: from bench to bedside. Eur J Pediatr 2014; 173:1123-36. [PMID: 24809802 DOI: 10.1007/s00431-014-2327-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/13/2014] [Accepted: 04/22/2014] [Indexed: 02/08/2023]
Abstract
UNLABELLED High-mobility group box protein 1 (HMGB1) is a nonhistone nuclear protein that has a dual function. Inside the cell, HMGB1 binds DNA, regulating transcription and determining chromosomal architecture. Outside the cell, HMGB1 activates the innate system and mediates a wide range of physiological and pathological responses. HMGB1 exerts these actions through differential engagement of multiple surface receptors, including Toll-like receptor (TLR)2, TLR4, and receptor for advanced glycation end products (RAGE). HMGB1 is implicated as a late mediator of sepsis and is also involved in inflammatory and autoimmune diseases, such as rheumatoid arthritis and systemic lupus erythematosus. Interestingly, HMGB1 was associated with tumor progression, becoming a potential therapeutic target, due to its involvement in the resistance to chemotherapy. Its implication on the pathogenesis of systemic vasculitis and inflammatory bowel diseases has also been evaluated. Moreover, it regulates neuroinflammation after traumatic brain injuries or cerebral infectious diseases. The aim of this review is to analyze these different roles of HMGB1, both in physiological and pathological conditions, discussing clinical and scientific implications in the field of pediatrics. CONCLUSION HMGB1 plays a key role in several pediatric diseases, opening new scenarios for diagnostic biomarkers and therapeutic strategies development.
Collapse
|
24
|
Valero N, Mosquera J, Levy A, Añez G, Marcucci R, Alvarez-Mon M. Differential induction of cytokines by human neonatal, adult, and elderly monocyte/macrophages infected with dengue virus. Viral Immunol 2014; 27:151-9. [PMID: 24801946 DOI: 10.1089/vim.2013.0123] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Immunosuppressive status against infections in monocytes from neonates and elderly subjects has been reported. The interaction between dengue virus and monocytes/macrophages plays an important role during dengue disease. The aim of this study was to determine the cytokine response of monocytes from individuals with different ages after infection with dengue virus. Monocyte/macrophage cultures from neonatal, adult, and elderly subjects (n=10 each group) were incubated with all four dengue virus types (DENV-1 to -4). After 1 and 3 days of culture, cytokine concentrations (TNF-α, IL-6, and IL-1β) were determined in culture supernatants by enzyme-linked immunosorbant assay. Increased production of all studied cytokines was induced by the different viral types in monocyte/macrophage cultures regardless of their source. However, lower cytokine concentrations were found in neonatal and elderly monocytes. The relative monocyte/macrophage immunosuppressive status observed in neonates and the elderly could be relevant during dengue infection in those age groups and important in innate and adaptive immunity responses against this virus.
Collapse
Affiliation(s)
- Nereida Valero
- 1 Instituto de Investigaciones Clínicas "Dr. Américo Negrette," Facultad de Medicina, Universidad del Zulia , Maracaibo, Venezuela
| | | | | | | | | | | |
Collapse
|
25
|
Qin Y, Chen Y, Wang W, Wang Z, Tang G, Zhang P, He Z, Liu Y, Dai SM, Shen Q. HMGB1-LPS complex promotes transformation of osteoarthritis synovial fibroblasts to a rheumatoid arthritis synovial fibroblast-like phenotype. Cell Death Dis 2014; 5:e1077. [PMID: 24556692 PMCID: PMC3944262 DOI: 10.1038/cddis.2014.48] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 01/18/2014] [Accepted: 01/20/2014] [Indexed: 12/29/2022]
Abstract
It is generally believed that some inflammatory antigens can recognize Toll-like receptors on synovial fibroblasts (SFs) and then activate downstream signals, leading to the formation of RASFs and inducing rheumatoid arthritis (RA). The objective of the current work was to study on the hypothesis that outer PAMP (LPS) binds to the inner DAMP (HMGB1) and becomes a complex that recognizes TLRs/RAGE on SFs, thus initiating a signaling cascade that leads to the secretion of inflammatory cytokines and chemokines, production of tissue-destructive enzymes, and formation of RASFs, finally resulting in RA. Osteoarthritis synovial fibroblasts (OASFs) were co-cultured with HMGB1–LPS complex in vitro for five generations to induce the transformation of human SFs to RA-like SFs (tOASFs). Then, changes of tOASFs in cell cycle and apoptosis–autophagy balance were investigated in vitro, and the pathogenicity of tOASFs was evaluated in a SCID mouse model in vivo. In vitro cell cycle analysis showed more tOASFs passing through the G1/S checkpoint and moving to S or G2 phase. Flow cytometry and confocal microscopy showed that apoptosis was reduced and autophagy was enhanced significantly in tOASFs as compared with those in OASFs. The expression of certain receptors and adhesion molecules in tOASFs was upregulated. In vivo experiments showed that tOASFs attached to, invaded, and degraded the co-implanted cartilage. In addition, histochemistry showed excessive proliferation of tOASFs and the expression of matrix metalloproteinases (MMPs). Based on the above findings, we conclude that HMGB1–LPS complex could promote the formation of RASFs.
Collapse
Affiliation(s)
- Y Qin
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - Y Chen
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - W Wang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai, China
| | - Z Wang
- Department of Orthopedics, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - G Tang
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - P Zhang
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - Z He
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - Y Liu
- Department of Rheumatology and Immunology, Changzheng Hospital of the Second Military Medical University, Shanghai, China
| | - S-M Dai
- Department of Rheumatology and Immunology, Changhai Hospital of the Second Military Medical University, Shanghai, China
| | - Q Shen
- Department of Laboratory Diagnosis, Changhai Hospital of the Second Military Medical University, Shanghai, China
| |
Collapse
|
26
|
Rojas A, Pérez-Castro R, González I, Delgado F, Romero J, Rojas I. The emerging role of the receptor for advanced glycation end products on innate immunity. Int Rev Immunol 2014; 33:67-80. [PMID: 24266871 DOI: 10.3109/08830185.2013.849702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2013] [Indexed: 02/05/2023]
Abstract
Cells from innate immune system are activated by the engagement of germ-line encoded pattern-recognition receptors (PRRs) in response to the microbial insult. These receptors are able to recognize either the presence of highly conserved microbial components called pathogen-associated molecular patterns or endogenous danger-associated molecular patterns. These danger signals are recognized by different types of (PRRs), including the receptor for advanced glycation end products. This new PRR share both ligands and intracellular signaling with Toll-like receptors and thus may cooperate with each other as essential partners to strength inflammatory response. This review summarizes recent advances in understanding the promiscuity of this receptor as well as its role in the context of innate immunity by triggering an inflammatory response when innate immune cells detect infection or tissue injury.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Laboratories, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | | | | | | | | | | |
Collapse
|
27
|
Yang C, Peng L, Su J. Two HMGB1 genes from grass carp Ctenopharyngodon idella mediate immune responses to viral/bacterial PAMPs and GCRV challenge. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 39:133-146. [PMID: 23228458 DOI: 10.1016/j.dci.2012.11.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 11/15/2012] [Accepted: 11/19/2012] [Indexed: 06/01/2023]
Abstract
High mobility group box 1 (HMGB1) is a nuclear weapon in the immune arsenal and a master regulator of innate immunity, at the crossroads between innate and adaptive immunity. To clarify the immune characterizations of HMGB1 in fishes, two co-orthologs of HMGB1 (CiHMGB1a and CiHMGB1b) were identified in grass carp Ctenopharyngodon idella by local EST database searching and RACE techniques. mRNA expressions of the two HMGB1 genes are widespread in fifteen tissues investigated. The transcripts of CiHMGB1a and CiHMGB1b were significantly up-regulated and reached peak at 24h post GCRV challenge in spleen and head kidney tissues (P<0.05). The modulations are slow post-bacterial PAMP stimulations by contrast with those after viral PAMP or GCRV challenge. They are inhibited by bacterial PAMPs, but are enhanced by viral PAMP or virus. mRNA expression of CiHMGB1a is high and strongly modulated by nucleic acids and transcription of CiHMGB1b is low and mildly regulated by nucleic acids and capsids of GCRV. The over-expression vectors were constructed and transfected into C. idella kidney cell line to obtain stably expressing recombinant proteins. In HMGB1 over-expressed cells, mRNA expressions of IPS-1, MyD88 and Mx1 were down-regulated, whereas TRIF was found to be up-regulated and IFN-I showed no change in its expression. After GCRV challenge, the transcripts of IPS-1, MyD88 and Mx1 were up-regulated, while IFN-I showed down-regulation, and TRIF showed up-regulation after an initial phase of decline. The titer assay demonstrated no antiviral activity of HMGB1s. The results indicated mRNA expressions of HMGB1a and HMGB1b are enhanced by GCRV or viral PAMP, and are inhibited by bacterial PAMPs; HMGB1a and HMGB1b collaborate with each other and play important roles in modulating the innate immune responses, although without direct antiviral effect; the immune network triggered by HMGB1 work together in concert to maintain homeostasis.
Collapse
Affiliation(s)
- Chunrong Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | | | | |
Collapse
|
28
|
Abstract
γδ-T cells represent a small population of immune cells, but play an indispensable role in host defenses against exogenous pathogens, immune surveillance of endogenous pathogenesis and even homeostasis of the immune system. Activation and expansion of γδ-T cells are generally observed in diverse human infectious diseases and correlate with their progression and prognosis. γδ-T cells have both 'innate' and 'adaptive' characteristics in the immune response, and their anti-infection activities are mediated by multiple pathways that are under elaborate regulation by other immune components. In this review, we summarize the current state of the literature and the recent advancements in γδ-T cell-mediated immune responses against common human infectious pathogens. Although further investigation is needed to improve our understanding of the characteristics of different γδ-T cell subpopulations under specific conditions, γδ-T cell-based therapy has great potential for the treatment of infectious diseases.
Collapse
|
29
|
Mo C, Dai Y, Kang N, Cui L, He W. Ectopic expression of human MutS homologue 2 on renal carcinoma cells is induced by oxidative stress with interleukin-18 promotion via p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) signaling pathways. J Biol Chem 2012; 287:19242-54. [PMID: 22493490 DOI: 10.1074/jbc.m112.349936] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human MutS homologue 2 (hMSH2), a crucial element of the highly conserved DNA mismatch repair system, maintains genetic stability in the nucleus of normal cells. Our previous studies indicate that hMSH2 is ectopically expressed on the surface of epithelial tumor cells and recognized by both T cell receptor γδ (TCRγδ) and natural killer group 2 member D (NKG2D) on Vδ2 T cells. Ectopically expressed hMSH2 could trigger a γδ T cell-mediated cytolysis. In this study, we showed that oxidative stress induced ectopic expression of hMSH2 on human renal carcinoma cells. Under oxidative stress, both p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK) pathways have been confirmed to mediate the ectopic expression of hMSH2 through the apoptosis-signaling kinase 1 (ASK1) upstream and activating transcription factor 3 (ATF3) downstream of both pathways. Moreover, renal carcinoma cell-derived interleukin (IL)-18 in oxidative stress was a prominent stimulator for ectopically induced expression of hMSH2, which was promoted by interferon (IFN)-γ as well. Finally, oxidative stress or pretreatment with IL-18 and IFN-γ enhanced γδ T cell-mediated cytolysis of renal carcinoma cells. Our results not only establish a mechanism of ectopic hMSH2 expression in tumor cells but also find a biological linkage between ectopic expression of hMSH2 and activation of γδ T cells in stressful conditions. Because γδ T cells play an important role in the early stage of innate anti-tumor response, γδ T cell activation triggered by ectopically expressed hMSH2 may be an important event in immunosurveillance for carcinogenesis.
Collapse
Affiliation(s)
- Chen Mo
- Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Peking Union Medical College, Beijing 100005, China
| | | | | | | | | |
Collapse
|