1
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [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: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
2
|
Alexander SN, Green AR, Debner EK, Ramos Freitas LE, Abdelhadi HMK, Szabo-Pardi TA, Burton MD. The influence of sex on neuroimmune communication, pain, and physiology. Biol Sex Differ 2024; 15:82. [PMID: 39439003 PMCID: PMC11494817 DOI: 10.1186/s13293-024-00660-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
With the National Institutes of Health's mandate to consider sex as a biological variable (SABV), there has been a significant increase of studies utilizing both sexes. Historically, we have known that biological sex and hormones influence immunological processes and now studies focusing on interactions between the immune, endocrine, and nervous systems are revealing sex differences that influence pain behavior and various molecular and biochemical processes. Neuroendocrine-immune interactions represent a key integrative discipline that will reveal critical processes in each field as it pertains to novel mechanisms in sex differences and necessary therapeutics. Here we appraise preclinical and clinical literature to discuss these interactions and key pathways that drive cell- and sex-specific differences in immunity, pain, and physiology.
Collapse
Affiliation(s)
- Shevon N Alexander
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Audrey R Green
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Emily K Debner
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Lindsey E Ramos Freitas
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Hanna M K Abdelhadi
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Thomas A Szabo-Pardi
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA
| | - Michael D Burton
- Neuroimmunology and Behavior Laboratory, Department of Neuroscience, School of Behavioral and Brain Sciences, Center for Advanced Pain Studies, University of Texas at Dallas, 800 W. Campbell Road, BSB 10.537, Richardson, TX, 75080, USA.
| |
Collapse
|
3
|
Lin CH, Tang LY, Wang LY, Chang CP. Thrombomodulin Improves Cognitive Deficits in Heat-Stressed Mice. Int J Neuropsychopharmacol 2024; 27:pyae027. [PMID: 38938182 PMCID: PMC11259854 DOI: 10.1093/ijnp/pyae027] [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: 10/20/2023] [Accepted: 06/24/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Thrombomodulin (TM) exerts anticoagulant and anti-inflammatory effects to improve the survival of patients with septic shock. Heat stroke resembles septic shock in many aspects. We tested whether TM would improve cognitive deficits and related causative factors in heat-stressed (HS) mice. METHODS Adult male mice were exposed to HS (33°C for 2 hours daily for 7 consecutive days) to induce cognitive deficits. Recombinant human soluble TM (1 mg/kg, i.p.) was administered immediately after the first HS trial and then once daily for 7 consecutive days. We performed the Y-maze, novel objective recognition, and passive avoidance tests to evaluate cognitive function. Plasma levels of lipopolysaccharide (LPS), high-mobility group box 1 (HMGB1), coagulation parameters, and both plasma and tissue levels of inflammatory and oxidative stress markers were biochemically measured. The duodenum and hippocampus sections were immunohistochemically stained. The intestinal and blood-brain barrier permeability were determined. RESULTS Compared with controls, HS mice treated with TM had lesser extents of cognitive deficits, exacerbated stress reactions, gut barrier disruption, endotoxemia, blood-brain barrier disruption, and inflammatory, oxidative, and coagulatory injury to heart, duodenum, and hippocampal tissues, and increased plasma HMGB1. In addition to reducing cognitive deficits, TM therapy alleviated all the abovementioned complications in heat-stressed mice. CONCLUSIONS The findings suggest that HS can lead to exacerbated stress reactions, endotoxemia, gut barrier disruption, blood-brain barrier disruption, hippocampal inflammation, coagulopathy, and oxidative stress, which may act as causative factors for cognitive deficits. TM, an anti-inflammatory, antioxidant, and anti-coagulatory agent, inhibited heat stress-induced cognitive deficits in mice.
Collapse
Affiliation(s)
- Cheng-Hsien Lin
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | | | - Lin-Yu Wang
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for General Education, Southern Taiwan University of Science and TechnologyTainan, Taiwan
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Pediatrics
- Chi Mei Medical Center, Tainan, Taiwan
| | | |
Collapse
|
4
|
Vásquez-Suárez A, Muñoz-Flores C, Ortega L, Roa F, Castillo C, Romero A, Parra N, Sandoval F, Macaya L, González-Chavarría I, Astuya A, Starck MF, Villegas MF, Agurto N, Montesino R, Sánchez O, Valenzuela A, Toledo JR, Acosta J. Design and functional characterization of Salmo salar TLR5 agonist peptides derived from high mobility group B1 acidic tail. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109373. [PMID: 38272332 DOI: 10.1016/j.fsi.2024.109373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Toll-like receptor 5 (TLR5) responds to the monomeric form of flagellin and induces the MyD88-depending signaling pathway, activating proinflammatory transcription factors such as NF-κB and the consequent induction of cytokines. On the other hand, HMGB1 is a highly conserved non-histone chromosomal protein shown to interact with and activate TLR5. The present work aimed to design and characterize TLR5 agonist peptides derived from the acidic tail of Salmo salar HMGB1 based on the structural knowledge of the TLR5 surface using global molecular docking platforms. Peptide binding poses complexed on TLR5 ectodomain model from each algorithm were filtrated based on docking scoring functions and predicted theoretical binding affinity of the complex. Circular dichroism spectra were recorded for each peptide selected for synthesis. Only intrinsically disordered peptides (6W, 11W, and SsOri) were selected for experimental functional assay. The functional characterization of the peptides was performed by NF-κB activation assays, RT-qPCR gene expression assays, and Piscirickettsia salmonis challenge in SHK-1 cells. The 6W and 11W peptides increased the nuclear translation of p65 and phosphorylation. In addition, the peptides induced the expression of genes related to the TLR5 pathway activation, pro- and anti-inflammatory response, and differentiation and activation of T lymphocytes towards phenotypes such as TH1, TH17, and TH2. Finally, it was shown that the 11W peptide protects immune cells against infection with P. salmonis bacteria. Overall, the results indicate the usefulness of novel peptides as potential immunostimulants in salmonids.
Collapse
Affiliation(s)
- Aleikar Vásquez-Suárez
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carolina Muñoz-Flores
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Leonardo Ortega
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Francisco Roa
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carolina Castillo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Alex Romero
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile; Centro FONDAP, Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
| | - Natalie Parra
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Sandoval
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Luis Macaya
- Departamento de Físico-Química, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Iván González-Chavarría
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Allisson Astuya
- Laboratorio de Genómica Marina y Cultivo Celular, Departamento de Oceanografía y COPAS Sur-Austral, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - María Francisca Starck
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Milton F Villegas
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Niza Agurto
- Laboratorio de Piscicultura y Patología Acuática, Departamento de Oceanografía, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Raquel Montesino
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Oliberto Sánchez
- Laboratorio de Biofármacos Recombinantes, Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ariel Valenzuela
- Laboratorio de Piscicultura y Patología Acuática, Departamento de Oceanografía, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Jorge R Toledo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - Jannel Acosta
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
5
|
Chen R, Zou J, Zhong X, Li J, Kang R, Tang D. HMGB1 in the interplay between autophagy and apoptosis in cancer. Cancer Lett 2024; 581:216494. [PMID: 38007142 DOI: 10.1016/j.canlet.2023.216494] [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: 07/21/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/27/2023]
Abstract
Lysosome-mediated autophagy and caspase-dependent apoptosis are dynamic processes that maintain cellular homeostasis, ensuring cell health and functionality. The intricate interplay and reciprocal regulation between autophagy and apoptosis are implicated in various human diseases, including cancer. High-mobility group box 1 (HMGB1), a nonhistone chromosomal protein, plays a pivotal role in coordinating autophagy and apoptosis levels during tumor initiation, progression, and therapy. The regulation of autophagy machinery and the apoptosis pathway by HMGB1 is influenced by various factors, including the protein's subcellular localization, oxidative state, and interactions with binding partners. In this narrative review, we provide a comprehensive overview of the structure and function of HMGB1, with a specific focus on the interplay between autophagic degradation and apoptotic death in tumorigenesis and cancer therapy. Gaining a comprehensive understanding of the significance of HMGB1 as a biomarker and its potential as a therapeutic target in tumor diseases is crucial for advancing our knowledge of cell survival and cell death.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Ju Zou
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Xiao Zhong
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jie Li
- Department of Infectious Diseases, Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
6
|
Li N, Han L, Wang X, Qiao O, Zhang L, Gong Y. Biotherapy of experimental acute kidney injury: emerging novel therapeutic strategies. Transl Res 2023; 261:69-85. [PMID: 37329950 DOI: 10.1016/j.trsl.2023.06.002] [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: 03/17/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Acute kidney injury (AKI) is a complex and heterogeneous disease with high incidence and mortality, posing a serious threat to human life and health. Usually, in clinical practice, AKI is caused by crush injury, nephrotoxin exposure, ischemia-reperfusion injury, or sepsis. Therefore, most AKI models for pharmacological experimentation are based on this. The current research promises to develop new biological therapies, including antibody therapy, non-antibody protein therapy, cell therapy, and RNA therapy, that could help mitigate the development of AKI. These approaches can promote renal repair and improve systemic hemodynamics after renal injury by reducing oxidative stress, inflammatory response, organelles damage, and cell death, or activating cytoprotective mechanisms. However, no candidate drugs for AKI prevention or treatment have been successfully translated from bench to bedside. This article summarizes the latest progress in AKI biotherapy, focusing on potential clinical targets and novel treatment strategies that merit further investigation in future pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Ning Li
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Lu Han
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Xinyue Wang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Li Zhang
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical College, Tianjin University, Nankai District, Tianjin, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, China.
| |
Collapse
|
7
|
Li J, Liu J, Zhou Z, Wu R, Chen X, Yu C, Stockwell B, Kroemer G, Kang R, Tang D. Tumor-specific GPX4 degradation enhances ferroptosis-initiated antitumor immune response in mouse models of pancreatic cancer. Sci Transl Med 2023; 15:eadg3049. [PMID: 37910602 DOI: 10.1126/scitranslmed.adg3049] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Abstract
Lipid peroxidation-dependent ferroptosis has become an emerging strategy for tumor therapy. However, current strategies not only selectively induce ferroptosis in malignant cells but also trigger ferroptosis in immune cells simultaneously, which can compromise anti-tumor immunity. Here, we used In-Cell Western assays combined with an unbiased drug screening to identify the compound N6F11 as a ferroptosis inducer that triggered the degradation of glutathione peroxidase 4 (GPX4), a key ferroptosis repressor, specifically in cancer cells. N6F11 did not cause the degradation of GPX4 in immune cells, including dendritic, T, natural killer, and neutrophil cells. Mechanistically, N6F11 bound to the RING domain of E3 ubiquitin ligase tripartite motif containing 25 (TRIM25) in cancer cells to trigger TRIM25-mediated K48-linked ubiquitination of GPX4, resulting in its proteasomal degradation. Functionally, N6F11 treatment caused ferroptotic cancer cell death that initiated HMGB1-dependent antitumor immunity mediated by CD8+ T cells. N6F11 also sensitized immune checkpoint blockade that targeted CD274/PD-L1 in advanced cancer models, including genetically engineered mouse models of pancreatic cancer driven by KRAS and TP53 mutations. These findings may establish a safe and efficient strategy to boost ferroptosis-driven antitumor immunity.
Collapse
Affiliation(s)
- Jingbo Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jiao Liu
- DAMP Laboratory, Third Affiliated Hospital, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, Guangzhou Medical University, Guangzhou, Guangdong 510510, China
| | - Zhuan Zhou
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Runliu Wu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xin Chen
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chunhua Yu
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Brent Stockwell
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris Cité, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, F-75015 Paris, France
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
8
|
Zheng X, Lu J, Liu J, Zhou L, He Y. HMGB family proteins: Potential biomarkers and mechanistic factors in cardiovascular diseases. Biomed Pharmacother 2023; 165:115118. [PMID: 37437373 DOI: 10.1016/j.biopha.2023.115118] [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: 05/05/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/14/2023] Open
Abstract
Cardiovascular disease (CVD) is the most fatal disease that causes sudden death, and inflammation contributes substantially to its occurrence and progression. The prevalence of CVD increases as the population ages, and the pathophysiology is complex. Anti-inflammatory and immunological modulation are the potential methods for CVD prevention and treatment. High-Mobility Group (HMG) chromosomal proteins are one of the most abundant nuclear nonhistone proteins which act as inflammatory mediators in DNA replication, transcription, and repair by producing cytokines and serving as damage-associated molecular patterns in inflammatory responses. The most common and well-studied HMG proteins are those with an HMGB domain, which participate in a variety of biological processes. HMGB1 and HMGB2 were the first members of the HMGB family to be identified and are present in all investigated eukaryotes. Our review is primarily concerned with the involvement of HMGB1 and HMGB2 in CVD. The purpose of this review is to provide a theoretical framework for diagnosing and treating CVD by discussing the structure and function of HMGB1 and HMGB2.
Collapse
Affiliation(s)
- Xialei Zheng
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Junmi Lu
- Department of Pathology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jing Liu
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Liufang Zhou
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Cardiovascular Medicine, the Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi 533000, China
| | - Yuhu He
- Department of Cardiology, the Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China.
| |
Collapse
|
9
|
Chen R, Zou J, Kang R, Tang D. The Redox Protein High-Mobility Group Box 1 in Cell Death and Cancer. Antioxid Redox Signal 2023; 39:569-590. [PMID: 36999916 DOI: 10.1089/ars.2023.0236] [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] [Indexed: 04/01/2023]
Abstract
Significance: As a redox-sensitive protein, high-mobility group box 1 (HMGB1) is implicated in regulating stress responses to oxidative damage and cell death, which are closely related to the pathology of inflammatory diseases, including cancer. Recent Advances: HMGB1 is a nonhistone nuclear protein that acts as a deoxyribonucleic acid chaperone to control chromosomal structure and function. HMGB1 can also be released into the extracellular space and function as a damage-associated molecular pattern protein during cell death, including during apoptosis, necrosis, necroptosis, pyroptosis, ferroptosis, alkaliptosis, and cuproptosis. Once released, HMGB1 binds to membrane receptors to shape immune and metabolic responses. In addition to subcellular localization, the function and activity of HMGB1 also depend on its redox state and protein posttranslational modifications. Abnormal HMGB1 plays a dual role in tumorigenesis and anticancer therapy (e.g., chemotherapy, radiation therapy, and immunotherapy) depending on the tumor types and stages. Critical Issues: A comprehensive understanding of the role of HMGB1 in cellular redox homeostasis is important for deciphering normal cellular functions and pathological manifestations. In this review, we discuss compartmental-defined roles of HMGB1 in regulating cell death and cancer. Understanding these advances may help us develop potential HMGB1-targeting drugs or approaches to treat oxidative stress-related diseases or pathological conditions. Future Directions: Further studies are required to dissect the mechanism by which HMGB1 maintains redox homeostasis under different stress conditions. A multidisciplinary effort is also required to evaluate the potential applications of precisely targeting the HMGB1 pathway in human health and disease. Antioxid. Redox Signal. 39, 569-590.
Collapse
Affiliation(s)
- Ruochan Chen
- Hunan Key Laboratory of Viral Hepatitis; Central South University, Changsha, China
- Department of Infectious Diseases; Xiangya Hospital, Central South University, Changsha, China
| | - Ju Zou
- Hunan Key Laboratory of Viral Hepatitis; Central South University, Changsha, China
- Department of Infectious Diseases; Xiangya Hospital, Central South University, Changsha, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
10
|
Zhang Y, Sheng Z, Xiao J, Li Y, Huang J, Jia J, Zeng X, Li L. Advances in the roles of glycyrrhizic acid in cancer therapy. Front Pharmacol 2023; 14:1265172. [PMID: 37649893 PMCID: PMC10463042 DOI: 10.3389/fphar.2023.1265172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 08/07/2023] [Indexed: 09/01/2023] Open
Abstract
Since the first 70 years of reporting cancer chemotherapy, malignant tumors have been the second most common cause of death in children and adults. Currently, the commonly used anti-cancer methods include surgery, chemotherapy, radiotherapy, and immunotherapy. Although these treatment methods could alleviate cancer, they lead to different forms of side effects and have no particularly significant effect on prolonging the patients' life span. Glycyrrhizic acid (GL), a native Chinese herbal extract, has a wide range of pharmacological effects, such as anti-cancer, anti-inflammatory, antioxidant, and immune regulation. In this review, the anti-cancer effects and mechanisms of GL are summarized in various cancers. The inhibition of GL on chemotherapy-induced side effects, including hepatotoxicity, nephrotoxicity, genotoxicity, neurotoxicity and pulmonary toxicity, is highlighted. Therefore, GL may be a promising and ideal drug for cancer therapy.
Collapse
Affiliation(s)
- Yuqian Zhang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Zixuan Sheng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jing Xiao
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Yang Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jie Huang
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
| | - Jinjing Jia
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Physiology, Jiaxing University Medical College, Jiaxing, China
| | - Xiansi Zeng
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Biochemistry and Molecular Biology, Jiaxing University Medical College, Jiaxing, China
| | - Li Li
- Research Center of Neuroscience, Jiaxing University Medical College, Jiaxing, China
- Department of Physiology, Jiaxing University Medical College, Jiaxing, China
| |
Collapse
|
11
|
Li J, Zhu CS, He L, Qiang X, Chen W, Wang H. A two-decade journey in identifying high mobility group box 1 (HMGB1) and procathepsin L (pCTS-L) as potential therapeutic targets for sepsis. Expert Opin Ther Targets 2023; 27:575-591. [PMID: 37477229 PMCID: PMC10530501 DOI: 10.1080/14728222.2023.2239495] [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: 02/20/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Microbial infections and resultant sepsis are leading causes of death in hospitals, representing approximately 20% of total deaths worldwide. Despite the difficulties in translating experimental insights into effective therapies for often heterogenous patient populations, an improved understanding of the pathogenic mechanisms underlying experimental sepsis is still urgently needed. Sepsis is partly attributable to dysregulated innate immune responses manifested by hyperinflammation and immunosuppression at different stages of microbial infections. AREAS COVERED Here we review our recent progress in searching for late-acting mediators of experimental sepsis and propose high mobility group box 1 (HMGB1) and procathepsin-L (pCTS-L) as potential therapeutic targets for improving outcomes of lethal sepsis and other infectious diseases. EXPERT OPINION It will be important to evaluate the efficacy of HMGB1- or pCTS-L-targeting agents for the clinical management of human sepsis and other infectious diseases in future studies.
Collapse
Affiliation(s)
- Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Cassie Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Li He
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| |
Collapse
|
12
|
Eek Mariampillai A, Hauge S, Kongsrud K, Syljuåsen RG. Immunogenic cell death after combined treatment with radiation and ATR inhibitors is dually regulated by apoptotic caspases. Front Immunol 2023; 14:1138920. [PMID: 37346039 PMCID: PMC10279842 DOI: 10.3389/fimmu.2023.1138920] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Introduction Inhibitors of the ATR kinase act as radiosensitizers through abrogating the G2 checkpoint and reducing DNA repair. Recent studies suggest that ATR inhibitors can also increase radiation-induced antitumor immunity, but the underlying immunomodulating mechanisms remain poorly understood. Moreover, it is poorly known how such immune effects relate to different death pathways such as caspase-dependent apoptosis. Here we address whether ATR inhibition in combination with irradiation may increase the presentation of hallmark factors of immunogenic cell death (ICD), and to what extent caspase activation regulates this response. Methods Human lung cancer and osteosarcoma cell lines (SW900, H1975, H460, U2OS) were treated with X-rays and ATR inhibitors (VE822; AZD6738) in the absence and presence of a pan-caspase inhibitor. The ICD hallmarks HMGB1 release, ATP secretion and calreticulin surface-presentation were assessed by immunoblotting of growth medium, the CellTiter-Glo assay and an optimized live-cell flow cytometry assay, respectively. To obtain accurate measurement of small differences in the calreticulin signal by flow cytometry, we included normalization to a barcoded control sample. Results Extracellular release of HMGB1 was increased in all the cell lines at 72 hours after the combined treatment with radiation and ATR inhibitors, relative to mock treatment or cells treated with radiation alone. The HMGB1 release correlated largely - but not strictly - with loss of plasma membrane integrity, and was suppressed by addition of the caspase inhibitor. However, one cell line showed HMGB1 release despite caspase inhibition, and in this cell line caspase inhibition induced pMLKL, a marker for necroptosis. ATP secretion occurred already at 48 hours after the co-treatment and did clearly not correlate with loss of plasma membrane integrity. Addition of pan-caspase inhibition further increased the ATP secretion. Surface-presentation of calreticulin was increased at 24-72 hours after irradiation, but not further increased by either ATR or caspase inhibition. Conclusion These results show that ATR inhibition can increase the presentation of two out of three ICD hallmark factors from irradiated human cancer cells. Moreover, caspase activation distinctly affects each of the hallmark factors, and therefore likely plays a dual role in tumor immunogenicity by promoting both immunostimulatory and -suppressive effects.
Collapse
Affiliation(s)
- Adrian Eek Mariampillai
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sissel Hauge
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Karoline Kongsrud
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Kwak MS, Choi S, Kim J, Lee H, Park IH, Oh J, Mai DN, Cho NH, Nam KT, Shin JS. SARS-CoV-2 Infection Induces HMGB1 Secretion Through Post-Translational Modification and PANoptosis. Immune Netw 2023; 23:e26. [PMID: 37416931 PMCID: PMC10320423 DOI: 10.4110/in.2023.23.e26] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/30/2023] [Accepted: 04/09/2023] [Indexed: 07/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection induces excessive pro-inflammatory cytokine release and cell death, leading to organ damage and mortality. High-mobility group box 1 (HMGB1) is one of the damage-associated molecular patterns that can be secreted by pro-inflammatory stimuli, including viral infections, and its excessive secretion levels are related to a variety of inflammatory diseases. Here, the aim of the study was to show that SARS-CoV-2 infection induced HMGB1 secretion via active and passive release. Active HMGB1 secretion was mediated by post-translational modifications, such as acetylation, phosphorylation, and oxidation in HEK293E/ACE2-C-GFP and Calu-3 cells during SARS-CoV-2 infection. Passive release of HMGB1 has been linked to various types of cell death; however, we demonstrated for the first time that PANoptosis, which integrates other cell death pathways, including pyroptosis, apoptosis, and necroptosis, is related to passive HMGB1 release during SARS-CoV-2 infection. In addition, cytoplasmic translocation and extracellular secretion or release of HMGB1 were confirmed via immunohistochemistry and immunofluorescence in the lung tissues of humans and angiotensin-converting enzyme 2-overexpressing mice infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Seoyeon Choi
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jiseon Kim
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Hoojung Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - In Ho Park
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jooyeon Oh
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Duong Ngoc Mai
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Department of Pediatrics, University of Medicine and Pharmacy, Ho Chi Minh 700000, Vietnam
| | - Nam-Hyuk Cho
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Ki Taek Nam
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
14
|
Chen W, Zhu CS, Qiang X, Chen S, Li J, Wang P, Tracey KJ, Wang H. Development of Procathepsin L (pCTS-L)-Inhibiting Lanosterol-Carrying Liposome Nanoparticles to Treat Lethal Sepsis. Int J Mol Sci 2023; 24:8649. [PMID: 37239992 PMCID: PMC10217857 DOI: 10.3390/ijms24108649] [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: 04/11/2023] [Revised: 05/03/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
The pathogenesis of microbial infections and sepsis is partly attributable to dysregulated innate immune responses propagated by late-acting proinflammatory mediators such as procathepsin L (pCTS-L). It was previously not known whether any natural product could inhibit pCTS-L-mediated inflammation or could be strategically developed into a potential sepsis therapy. Here, we report that systemic screening of a NatProduct Collection of 800 natural products led to the identification of a lipophilic sterol, lanosterol (LAN), as a selective inhibitor of pCTS-L-induced production of cytokines [e.g., Tumor Necrosis Factor (TNF) and Interleukin-6 (IL-6)] and chemokines [e.g., Monocyte Chemoattractant Protein-1 (MCP-1) and Epithelial Neutrophil-Activating Peptide (ENA-78)] in innate immune cells. To improve its bioavailability, we generated LAN-carrying liposome nanoparticles and found that these LAN-containing liposomes (LAN-L) similarly inhibited pCTS-L-induced production of several chemokines [e.g., MCP-1, Regulated upon Activation, Normal T Cell Expressed and Presumably Secreted (RANTES) and Macrophage Inflammatory Protein-2 (MIP-2)] in human blood mononuclear cells (PBMCs). In vivo, these LAN-carrying liposomes effectively rescued mice from lethal sepsis even when the first dose was given at 24 h post the onset of this disease. This protection was associated with a significant attenuation of sepsis-induced tissue injury and systemic accumulation of serval surrogate biomarkers [e.g., IL-6, Keratinocyte-derived Chemokine (KC), and Soluble Tumor Necrosis Factor Receptor I (sTNFRI)]. These findings support an exciting possibility to develop liposome nanoparticles carrying anti-inflammatory sterols as potential therapies for human sepsis and other inflammatory diseases.
Collapse
Affiliation(s)
- Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| | - Cassie Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| | - Shujin Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
| | - Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
| | - Ping Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| | - Kevin J. Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, New York, NY 11030, USA; (W.C.); (C.S.Z.); (X.Q.); (S.C.); (J.L.); (P.W.); (K.J.T.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, New York, NY 11549, USA
| |
Collapse
|
15
|
Mo Y, Chen K. Review: The role of HMGB1 in spinal cord injury. Front Immunol 2023; 13:1094925. [PMID: 36713448 PMCID: PMC9877301 DOI: 10.3389/fimmu.2022.1094925] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
High mobility group box 1 (HMGB1) has dual functions as a nonhistone nucleoprotein and an extracellular inflammatory cytokine. In the resting state, HMGB1 is mainly located in the nucleus and regulates key nuclear activities. After spinal cord injury, HMGB1 is rapidly expressed by neurons, microglia and ependymal cells, and it is either actively or passively released into the extracellular matrix and blood circulation; furthermore, it also participates in the pathophysiological process of spinal cord injury. HMGB1 can regulate the activation of M1 microglia, exacerbate the inflammatory response, and regulate the expression of inflammatory factors through Rage and TLR2/4, resulting in neuronal death. However, some studies have shown that HMGB1 is beneficial for the survival, regeneration and differentiation of neurons and that it promotes the recovery of motor function. This article reviews the specific timing of secretion and translocation, the release mechanism and the role of HMGB1 in spinal cord injury. Furthermore, the role and mechanism of HMGB1 in spinal cord injury and, the challenges that still need to be addressed are identified, and this work will provide a basis for future studies.
Collapse
|
16
|
Schmitt M, Ceteci F, Gupta J, Pesic M, Böttger TW, Nicolas AM, Kennel KB, Engel E, Schewe M, Callak Kirisözü A, Petrocelli V, Dabiri Y, Varga J, Ramakrishnan M, Karimova M, Ablasser A, Sato T, Arkan MC, de Sauvage FJ, Greten FR. Colon tumour cell death causes mTOR dependence by paracrine P2X4 stimulation. Nature 2022; 612:347-353. [PMID: 36385525 PMCID: PMC7613947 DOI: 10.1038/s41586-022-05426-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 10/07/2022] [Indexed: 11/17/2022]
Abstract
Solid cancers exhibit a dynamic balance between cell death and proliferation ensuring continuous tumour maintenance and growth1,2. Increasing evidence links enhanced cancer cell apoptosis to paracrine activation of cells in the tumour microenvironment initiating tissue repair programs that support tumour growth3,4, yet the direct effects of dying cancer cells on neighbouring tumour epithelia and how this paracrine effect potentially contributes to therapy resistance are unclear. Here we demonstrate that chemotherapy-induced tumour cell death in patient-derived colorectal tumour organoids causes ATP release triggering P2X4 (also known as P2RX4) to mediate an mTOR-dependent pro-survival program in neighbouring cancer cells, which renders surviving tumour epithelia sensitive to mTOR inhibition. The induced mTOR addiction in persisting epithelial cells is due to elevated production of reactive oxygen species and subsequent increased DNA damage in response to the death of neighbouring cells. Accordingly, inhibition of the P2X4 receptor or direct mTOR blockade prevents induction of S6 phosphorylation and synergizes with chemotherapy to cause massive cell death induced by reactive oxygen species and marked tumour regression that is not seen when individually applied. Conversely, scavenging of reactive oxygen species prevents cancer cells from becoming reliant on mTOR activation. Collectively, our findings show that dying cancer cells establish a new dependency on anti-apoptotic programs in their surviving neighbours, thereby creating an opportunity for combination therapy in P2X4-expressing epithelial tumours.
Collapse
Affiliation(s)
- Mark Schmitt
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
- Institute of Pharmacology, University of Marburg, Marburg, Germany
| | - Fatih Ceteci
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jalaj Gupta
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
- Stem Cell Research Center, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Marina Pesic
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Tim W Böttger
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Adele M Nicolas
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Kilian B Kennel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Esther Engel
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Matthias Schewe
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Asude Callak Kirisözü
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Valentina Petrocelli
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Yasamin Dabiri
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Julia Varga
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Mallika Ramakrishnan
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Madina Karimova
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Andrea Ablasser
- Global Health Institute, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Melek C Arkan
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany
| | | | - Florian R Greten
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany.
- Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
17
|
High Mobility Group Box-1 regulates expression of EGFR, VEGF, StAR and TIMP1/2 in bovine granulosa cells through a mechanism involving TLR2/NF-κB. Anim Reprod Sci 2022; 247:107152. [DOI: 10.1016/j.anireprosci.2022.107152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/08/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
|
18
|
Wu E, Zhu J, Ma Z, Tuo B, Terai S, Mizuno K, Li T, Liu X. Gastric alarmin release: A warning signal in the development of gastric mucosal diseases. Front Immunol 2022; 13:1008047. [PMID: 36275647 PMCID: PMC9583272 DOI: 10.3389/fimmu.2022.1008047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 09/14/2022] [Indexed: 11/13/2022] Open
Abstract
Alarmins exist outside cells and are early warning signals to the immune system; as such, alarmin receptors are widely distributed on various immune cells. Alarmins, proinflammatory molecular patterns associated with tissue damage, are usually released into the extracellular space, where they induce immune responses and participate in the damage and repair processes of mucosal diseases.In the stomach, gastric alarmin release has been shown to be involved in gastric mucosal inflammation, antibacterial defense, adaptive immunity, and wound healing; moreover, this release causes damage and results in the development of gastric mucosal diseases, including various types of gastritis, ulcers, and gastric cancer. Therefore, it is necessary to understand the role of alarmins in gastric mucosal diseases. This review focuses on the contribution of alarmins, including IL33, HMGB1, defensins and cathelicidins, to the gastric mucosal barrier and their role in gastric mucosal diseases. Here, we offer a new perspective on the prevention and treatment of gastric mucosal diseases.
Collapse
Affiliation(s)
- Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiaxing Zhu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenichi Mizuno
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- *Correspondence: Xuemei Liu, ; Taolang Li,
| |
Collapse
|
19
|
Yang R, Zhang X. A potential new pathway for heparin treatment of sepsis-induced lung injury: inhibition of pulmonary endothelial cell pyroptosis by blocking hMGB1-LPS-induced caspase-11 activation. Front Cell Infect Microbiol 2022; 12:984835. [PMID: 36189354 PMCID: PMC9519888 DOI: 10.3389/fcimb.2022.984835] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022] Open
Abstract
Sepsis is a significant cause of mortality in critically ill patients. Acute lung injury (ALI) is a leading cause of death in these patients. Endothelial cells exposed to the bacterial endotoxin lipopolysaccharide (LPS) can progress into pyroptosis, a programmed lysis of cell death triggered by inflammatory caspases. It is characterized by lytic cell death induced by the binding of intracellular LPS to caspases 4/5 in human cells and caspase-11 in mouse cells. In mice,caspase-11-dependent pyroptosis plays an important role in endotoxemia. HMGB1 released into the plasma binds to LPS and is internalized into lysosomes in endothelial cells via the advanced glycation end product receptor. In the acidic lysosomal environment, HMGB1 permeates the phospholipid bilayer, which is followed by the leakage of LPS into the cytoplasm and the activation of caspase-11. Heparin is an anticoagulant widely applied in the treatment of thrombotic disease. Previous studies have found that heparin could block caspase-11-dependent inflammatory reactions, decrease sepsis-related mortality, and reduce ALI, independent of its anticoagulant activity. Heparin or modified heparin with no anticoagulant property could inhibit the alarmin HMGB1-LPS interactions, minimize LPS entry into the cytoplasm, and thus blocking caspase-11 activation. Heparin has been studied in septic ALI, but the regulatory mechanism of pulmonary endothelial cell pyroptosis is still unclear. In this paper, we discuss the potential novel role of heparin in the treatment of septic ALI from the unique mechanism of pulmonary endothelial cell pyroptosis.
Collapse
|
20
|
Deng C, Deng L, Lv J, Sun L. Therapeutic effects and long-term outcomes of HMGB1-targeted therapy in rats and mice with traumatic spinal cord injury: A systematic review and meta-analysis. Front Neurosci 2022; 16:968791. [PMID: 36161176 PMCID: PMC9489835 DOI: 10.3389/fnins.2022.968791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/18/2022] [Indexed: 12/09/2022] Open
Abstract
BackgroundTo date, the clinical need for therapeutic methods to prevent traumatic spinal cord injury (TSCI) progression and improve functional recovery has not been met. High mobility group box-1 (HMGB1) is released by necrotic neurons or secreted by glial cells after TSCI and plays an important role in pathophysiology.ObjectiveThe purpose of this study was to evaluate the effects of HMGB1-targeted therapy on locomotor function recovery, inflammation reduction, edema attenuation, and apoptosis reduction in rat and mouse models of TSCI.MethodsWe reviewed the literature on HMGB1-targeted therapy in the treatment and prognosis of TSCI. Twelve articles were identified and analyzed from four online databases (PubMed, Web of Science, Cochrane Library and Embase) based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines and strict inclusion criteria.ResultsThe methodological quality of the 12 articles was poor. The results of the meta-analysis showed that compared with the SCI group, the treatment group had significantly increased locomotor function scores after SCI [n = 159, standardized mean difference (SMD) = 2.31, 95% confidence interval (CI) (1.52, 3.10), P < 0.00001], and the change in locomotor function scores was significantly increased in both the drug and anti-HMGB1 Ab groups (P < 0.000001 and P < 0.000001). A subgroup analysis showed significant differences (P > 0.05) between the drug group [(SMD) = 1.95, 95% CI (0.95, 2.94), P = 0.0001] and the anti-HMGB1 Ab group [(SMD) = 2.89, 95% CI (1.66, 4.13), P < 0.00001]. Compared with the SCI group, HMGB1 expression was significantly diminished [n = 76, SMD = −2.31, 95% CI (−3.71, −0.91), P = 0.001], TNF-α levels were significantly reduced [n = 76, SMD = −2.52, 95% CI (−3.77, −1.27), P < 0.0001], water content was significantly reduced [n = 44, SMD = −3.94, 95% CI (−6.28, −1.61), P = 0.0009], and the number of apoptotic cells was significantly diminished [n = 36, SMD = −3.31, 95% CI (−6.40, −0.22), P = 0.04] in the spinal cord of the treatment group.ConclusionHMGB1-targeted therapy improves locomotor function, reduces inflammation, attenuates edema, and reduces apoptosis in rats and mice with TSCI. Intrathecal injection of anti-HMGB1 Ab 0-3 h after SCI may be the most efficacious treatment.Systematic review registrationPROSPERO, identifier: CRD42022326114.
Collapse
|
21
|
Chen Y, Yu C, Jiang S, Sun L. Japanese Flounder HMGB1: A DAMP Molecule That Promotes Antimicrobial Immunity by Interacting with Immune Cells and Bacterial Pathogen. Genes (Basel) 2022; 13:genes13091509. [PMID: 36140677 PMCID: PMC9498587 DOI: 10.3390/genes13091509] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/16/2022] Open
Abstract
High mobility group box (HMGB) proteins are DNA-associated proteins that bind and modulate chromosome structures. In mammals, HMGB proteins can be released from the cell nucleus and serve as a damage-associated molecular pattern (DAMP) under stress conditions. In fish, the DAMP function of HMGB proteins in association with bacterial infection remains to be investigated. In this study, we examined the immunological functions of two HMGB members, HMGB1 and HMG20A, of Japanese flounder. HMGB1 and HMG20A were expressed in multiple tissues of the flounder. HMGB1 was released from peripheral blood leukocytes (PBLs) upon bacterial challenge in a temporal manner similar to that of lactate dehydrogenase release. Recombinant HMGB1 bound to PBLs and induced ROS production and the expression of inflammatory genes. HMGB1 as well as HMG20A also bound to various bacterial pathogens and caused bacterial agglutination. The bacteria-binding patterns of HMGB1 and HMG20A were similar, and the binding of HMGB1 competed with the binding of HMG20A but not vice versa. During bacterial infection, HMGB1 enhanced the immune response of PBLs and repressed bacterial invasion. Collectively, our results indicate that flounder HMGB1 plays an important role in antimicrobial immunity by acting both as a modulator of immune cells and as a pathogen-interacting DAMP.
Collapse
Affiliation(s)
- Yuan Chen
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chao Yu
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Jiang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Sun
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, China
- Laboratory for Marine Biology and Biotechnology, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- College of Earth and Planetary Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- Correspondence:
| |
Collapse
|
22
|
Nofi CP, Wang P, Aziz M. Chromatin-Associated Molecular Patterns (CAMPs) in sepsis. Cell Death Dis 2022; 13:700. [PMID: 35961978 PMCID: PMC9372964 DOI: 10.1038/s41419-022-05155-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/28/2022] [Accepted: 08/01/2022] [Indexed: 01/21/2023]
Abstract
Several molecular patterns have been identified that recognize pattern recognition receptors. Pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs) are commonly used terminologies to classify molecules originating from pathogen and endogenous molecules, respectively, to heighten the immune response in sepsis. Herein, we focus on a subgroup of endogenous molecules that may be detected as foreign and similarly trigger immune signaling pathways. These chromatin-associated molecules, i.e., chromatin containing nuclear DNA and histones, extracellular RNA, mitochondrial DNA, telomeric repeat-containing RNA, DNA- or RNA-binding proteins, and extracellular traps, may be newly classified as chromatin-associated molecular patterns (CAMPs). Herein, we review the release of CAMPs from cells, their mechanism of action and downstream immune signaling pathways, and targeted therapeutic approaches to mitigate inflammation and tissue injury in inflammation and sepsis.
Collapse
Affiliation(s)
- Colleen P. Nofi
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| | - Ping Wang
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| | - Monowar Aziz
- grid.250903.d0000 0000 9566 0634Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, Manhasset, NY USA ,Elmezi Graduate School of Molecular Medicine, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Surgery, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA ,grid.512756.20000 0004 0370 4759Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY USA
| |
Collapse
|
23
|
Abstract
Sepsis, a systemic inflammatory response disease, is the most severe complication of infection and a deadly disease. High mobility group proteins (HMGs) are non-histone nuclear proteins binding nucleosomes and regulate chromosome architecture and gene transcription, which act as a potent pro-inflammatory cytokine involved in the delayed endotoxin lethality and systemic inflammatory response. HMGs increase in serum and tissues during infection, especially in sepsis. A growing number of studies have demonstrated HMGs are not only cytokines which can mediate inflammation, but also potential therapeutic targets in sepsis. To reduce sepsis-related mortality, a better understanding of HMGs is essential. In this review, we described the structure and function of HMGs, summarized the definition, epidemiology and pathophysiology of sepsis, and discussed the HMGs-related mechanisms in sepsis from the perspectives of non-coding RNAs (microRNA, long non-coding RNA, circular RNA), programmed cell death (apoptosis, necroptosis and pyroptosis), drugs and other pathophysiological aspects to provide new targets and ideas for the diagnosis and treatment of sepsis.
Collapse
Affiliation(s)
- Guibin Liang
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhihui He
- Department of Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
24
|
Dulmovits BM, Tang Y, Papoin J, He M, Li J, Yang H, Addorisio ME, Kennedy L, Khan M, Brindley E, Ashley RJ, Ackert-Bicknell C, Hale J, Kurita R, Nakamura Y, Diamond B, Barnes BJ, Hermine O, Gallagher PG, Steiner LA, Lipton JM, Taylor N, Mohandas N, Andersson U, Al-Abed Y, Tracey KJ, Blanc L. HMGB1-mediated restriction of EPO signaling contributes to anemia of inflammation. Blood 2022; 139:3181-3193. [PMID: 35040907 PMCID: PMC9136881 DOI: 10.1182/blood.2021012048] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 12/22/2021] [Indexed: 11/20/2022] Open
Abstract
Anemia of inflammation, also known as anemia of chronic disease, is refractory to erythropoietin (EPO) treatment, but the mechanisms underlying the EPO refractory state are unclear. Here, we demonstrate that high mobility group box-1 protein (HMGB1), a damage-associated molecular pattern molecule recently implicated in anemia development during sepsis, leads to reduced expansion and increased death of EPO-sensitive erythroid precursors in human models of erythropoiesis. HMGB1 significantly attenuates EPO-mediated phosphorylation of the Janus kinase 2/STAT5 and mTOR signaling pathways. Genetic ablation of receptor for advanced glycation end products, the only known HMGB1 receptor expressed by erythroid precursors, does not rescue the deleterious effects of HMGB1 on EPO signaling, either in human or murine precursors. Furthermore, surface plasmon resonance studies highlight the ability of HMGB1 to interfere with the binding between EPO and the EPOR. Administration of a monoclonal anti-HMGB1 antibody after sepsis onset in mice partially restores EPO signaling in vivo. Thus, HMGB1-mediated restriction of EPO signaling contributes to the chronic phase of anemia of inflammation.
Collapse
Affiliation(s)
- Brian M Dulmovits
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | | | | | - Mingzhu He
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Jianhua Li
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Huan Yang
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Meghan E Addorisio
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | | | | | - Elena Brindley
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Ryan J Ashley
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | | | - John Hale
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY
| | - Ryo Kurita
- Central Blood Institute, Japanese Red Cross Society, Minato-ku, Tokyo, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Research Center, Tsukuba, Ibaraki, Japan
| | - Betty Diamond
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Betsy J Barnes
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
| | - Olivier Hermine
- INSERM Unité Mixte de Recherche (UMR) 1163, IMAGINE Institute, Paris, France
| | | | - Laurie A Steiner
- Department of Pediatrics, University of Rochester, Rochester, NY
| | - Jeffrey M Lipton
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
- Pediatric Hematology/Oncology, Cohen Children's Medical Center, New Hyde Park, NY
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, Center for Cancer Research, National Institutes of Health, Bethesda, MD; and
| | - Narla Mohandas
- Red Cell Physiology Laboratory, New York Blood Center, New York, NY
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Yousef Al-Abed
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Kevin J Tracey
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY
| | - Lionel Blanc
- Zucker School of Medicine at Hofstra Northwell, Hempstead, NY
- Institute of Molecular Medicine, and
- INSERM Unité Mixte de Recherche (UMR) 1163, IMAGINE Institute, Paris, France
| |
Collapse
|
25
|
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: 3.5] [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
|
26
|
Bhat SM, Massey N, Shrestha D, Karriker LA, Jelesijević T, Wang C, Charavaryamath C. Transcriptomic and ultrastructural evidence indicate that anti-HMGB1 antibodies rescue organic dust-induced mitochondrial dysfunction. Cell Tissue Res 2022; 388:373-398. [PMID: 35244775 PMCID: PMC10155187 DOI: 10.1007/s00441-022-03602-3] [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: 08/04/2021] [Accepted: 02/22/2022] [Indexed: 11/02/2022]
Abstract
Exposure to organic dust (OD) in agriculture is known to cause respiratory symptoms including loss of lung function. OD exposure activates multiple signaling pathways since it contains a variety of microbial products and particulate matter. Previously, we have shown how OD exposure leads to the secretion of HMGB1 and HMGB1-RAGE signaling, and how this can be a possible therapeutic target to reduce inflammation. Cellular mitochondria are indispensable for homeostasis and are emerging targets to curtail inflammation. Recently, we have also observed that OD exposure induces mitochondrial dysfunction characterized by loss of structural integrity and deficits in bioenergetics. However, the role of HMGB1 in OD-induced mitochondrial dysfunction in human bronchial epithelial (NHBE) cells remains elusive. Therefore, we aimed to study whether decreased levels of intracellular HMGB1 or antibody-mediated neutralization of secreted HMGB1 would rescue mitochondrial dysfunction. Single and repeated ODE exposure showed an elongated mitochondrial network and cristolysis whereas HMGB1 neutralization or the lack thereof promotes mitochondrial biogenesis evidenced by increased mitochondrial fragmentation, increased DRP1 expression, decreased MFN2 expression, and increased PGC1α expression. Repeated 5-day ODE exposure significantly downregulated transcripts encoding mitochondrial respiration and metabolism (ATP synthase, NADUF, and UQCR) as well as glucose uptake. This was reversed by the antibody-mediated neutralization of HMGB1. Our results support our hypothesis that, in NHBE cells, neutralization of ODE-induced HMGB1 secretion rescues OD-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Sanjana Mahadev Bhat
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
- Immunobiology Interdepartmental Graduate Program, Iowa State University, Ames, IA, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Nyzil Massey
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Denusha Shrestha
- Department of Biomedical Sciences, Iowa State University, Ames, IA, USA
| | - Locke A Karriker
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, USA
| | - Tomislav Jelesijević
- Department of Comparative Biological Sciences, Louisiana State University, Baton Rouge, LA, USA
| | - Chong Wang
- Department of Veterinary Diagnostic and Production Animal Medicine, Iowa State University, Ames, IA, USA
- Department of Statistics, Iowa State University, Ames, IA, USA
| | | |
Collapse
|
27
|
Knijff LWD, van Kooten C, Ploeg RJ. The Effect of Hypothermic Machine Perfusion to Ameliorate Ischemia-Reperfusion Injury in Donor Organs. Front Immunol 2022; 13:848352. [PMID: 35572574 PMCID: PMC9099247 DOI: 10.3389/fimmu.2022.848352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/04/2022] [Indexed: 12/23/2022] Open
Abstract
Hypothermic machine perfusion (HMP) has become the new gold standard in clinical donor kidney preservation and a promising novel strategy in higher risk donor livers in several countries. As shown by meta-analysis for the kidney, HMP decreases the risk of delayed graft function (DGF) and improves graft survival. For the liver, HMP immediately prior to transplantation may reduce the chance of early allograft dysfunction (EAD) and reduce ischemic sequelae in the biliary tract. Ischemia-reperfusion injury (IRI), unavoidable during transplantation, can lead to massive cell death and is one of the main causes for DGF, EAD or longer term impact. Molecular mechanisms that are affected in IRI include levels of hypoxia inducible factor (HIF), induction of cell death, endothelial dysfunction and immune responses. In this review we have summarized and discussed mechanisms on how HMP can ameliorate IRI. Better insight into how HMP influences IRI in kidney and liver transplantation may lead to new therapies and improved transplant outcomes.
Collapse
Affiliation(s)
- Laura W. D. Knijff
- Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, Netherlands
- Transplant Centre of the Leiden University Medical Centre, Leiden University Medical Centre, Leiden, Netherlands
| | - Cees van Kooten
- Nephrology, Department of Internal Medicine, Leiden University Medical Centre, Leiden, Netherlands
- Transplant Centre of the Leiden University Medical Centre, Leiden University Medical Centre, Leiden, Netherlands
| | - Rutger J. Ploeg
- Transplant Centre of the Leiden University Medical Centre, Leiden University Medical Centre, Leiden, Netherlands
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
High Mobility Group Box 1: Biological Functions and Relevance in Oxidative Stress Related Chronic Diseases. Cells 2022; 11:cells11050849. [PMID: 35269471 PMCID: PMC8909428 DOI: 10.3390/cells11050849] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/03/2022] [Accepted: 02/26/2022] [Indexed: 01/27/2023] Open
Abstract
In the early 1970s, a group of non-histone nuclear proteins with high electrophoretic mobility was discovered and named high-mobility group (HMG) proteins. High-mobility group box 1 (HMGB1) is the most studied HMG protein that detects and coordinates cellular stress response. The biological function of HMGB1 depends on its subcellular localization and expression. It plays a critical role in the nucleus and cytoplasm as DNA chaperone, chromosome gatekeeper, autophagy maintainer, and protector from apoptotic cell death. HMGB1 also functions as an extracellular alarmin acting as a damage-associated molecular pattern molecule (DAMP). Recent findings describe HMGB1 as a sophisticated signal of danger, with a pleiotropic function, which is useful as a clinical biomarker for several disorders. HMGB1 has emerged as a mediator in acute and chronic inflammation. Furthermore, HMGB1 targeting can induce beneficial effects on oxidative stress related diseases. This review focus on HMGB1 redox status, localization, mechanisms of release, binding with receptors, and its activities in different oxidative stress-related chronic diseases. Since a growing number of reports show the key role of HMGB1 in socially relevant pathological conditions, to our knowledge, for the first time, here we analyze the scientific literature, evaluating the number of publications focusing on HMGB1 in humans and animal models, per year, from 2006 to 2021 and the number of records published, yearly, per disease and category (studies on humans and animal models).
Collapse
|
29
|
Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022; 54:91-102. [PMID: 35217834 PMCID: PMC8894452 DOI: 10.1038/s12276-022-00736-w] [Citation(s) in RCA: 291] [Impact Index Per Article: 145.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/13/2021] [Accepted: 11/04/2021] [Indexed: 02/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a nonhistone nuclear protein that has multiple functions according to its subcellular location. In the nucleus, HMGB1 is a DNA chaperone that maintains the structure and function of chromosomes. In the cytoplasm, HMGB1 can promote autophagy by binding to BECN1 protein. After its active secretion or passive release, extracellular HMGB1 usually acts as a damage-associated molecular pattern (DAMP) molecule, regulating inflammation and immune responses through different receptors or direct uptake. The secretion and release of HMGB1 is fine-tuned by a variety of factors, including its posttranslational modification (e.g., acetylation, ADP-ribosylation, phosphorylation, and methylation) and the molecular machinery of cell death (e.g., apoptosis, pyroptosis, necroptosis, alkaliptosis, and ferroptosis). In this minireview, we introduce the basic structure and function of HMGB1 and focus on the regulatory mechanism of HMGB1 secretion and release. Understanding these topics may help us develop new HMGB1-targeted drugs for various conditions, especially inflammatory diseases and tissue damage. A nuclear protein that gets released after cell death or is actively secreted by immune cells offers a promising therapeutic target for treating diseases linked to excessive inflammation. Daolin Tang from the University of Texas Southwestern Medical Center in Dallas, USA, and colleagues review how cellular stresses can trigger the accumulation of HMGB1, a type of alarm signal protein that promotes the recruitment and activation of inflammation-promoting immune cells. The researchers discuss various mechanisms that drive both passive and active release of HMGB1 into the space around cells. These processes, which include enzymatic modifications of the HMGB1 protein, cell–cell interactions and molecular pathways of cell death, could be targeted by drugs to lessen tissue damage and inflammatory disease caused by HMGB1-induced immune responses
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China. .,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
30
|
Chen H, Agrawal DK, Thankam FG. Biomaterials-Driven Sterile Inflammation. TISSUE ENGINEERING. PART B, REVIEWS 2022; 28:22-34. [PMID: 33213285 PMCID: PMC8892963 DOI: 10.1089/ten.teb.2020.0253] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Performance of the biomaterials used for regenerative medicine largely depends on biocompatibility; however, the biological mechanisms underlying biocompatibility of a biomaterial within the host system is poorly understood. In addition to the classical immune response against non-self-entities, the sterile inflammatory response could limit the compatibility of biological scaffolds. Whereas the immediate to short-term host response to a biomaterial implant have been characterized, the long-term progression of host-biomaterial relationship has not been described. This article explores the novel concept of biomaterials-driven sterile inflammation (BSI) in long-term biodegradable implants and throws light for possible explanation for the onset of BSI and the associated damage-associated molecular patterns. The understanding of BSI would advance the current strategies to improve biomaterial-host tissue integration and open novel translational avenues in biomaterials-based tissue regeneration. Impact statement Understanding the novel concept of biomaterials-driven sterile inflammation and associated damage-associated molecular patterns in long-term biodegradable implants would determine their success and improves the tissue engineering and regenerative strategies.
Collapse
Affiliation(s)
- Henry Chen
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Devendra K. Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
| | - Finosh G. Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California, USA
- Address correspondence to: Finosh G. Thankam, PhD, Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA 91766-1854, USA
| |
Collapse
|
31
|
Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther 2021; 6:407. [PMID: 34824200 PMCID: PMC8613465 DOI: 10.1038/s41392-021-00816-9] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/19/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection. Over decades, advanced understanding of host-microorganism interaction has gradually unmasked the genuine nature of sepsis, guiding toward new definition and novel therapeutic approaches. Diverse clinical manifestations and outcomes among infectious patients have suggested the heterogeneity of immunopathology, while systemic inflammatory responses and deteriorating organ function observed in critically ill patients imply the extensively hyperactivated cascades by the host defense system. From focusing on microorganism pathogenicity, research interests have turned toward the molecular basis of host responses. Though progress has been made regarding recognition and management of clinical sepsis, incidence and mortality rate remain high. Furthermore, clinical trials of therapeutics have failed to obtain promising results. As far as we know, there was no systematic review addressing sepsis-related molecular signaling pathways and intervention therapy in literature. Increasing studies have succeeded to confirm novel functions of involved signaling pathways and comment on efficacy of intervention therapies amid sepsis. However, few of these studies attempt to elucidate the underlining mechanism in progression of sepsis, while other failed to integrate preliminary findings and describe in a broader view. This review focuses on the important signaling pathways, potential molecular mechanism, and pathway-associated therapy in sepsis. Host-derived molecules interacting with activated cells possess pivotal role for sepsis pathogenesis by dynamic regulation of signaling pathways. Cross-talk and functions of these molecules are also discussed in detail. Lastly, potential novel therapeutic strategies precisely targeting on signaling pathways and molecules are mentioned.
Collapse
Affiliation(s)
- Yun-Yu Zhang
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China
| | - Bo-Tao Ning
- Department of Pediatric Intensive Care Unit, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 200127, Shanghai, China.
| |
Collapse
|
32
|
Wang S, Dong J, Li L, Wu R, Xu L, Ren Y, Hu X. Exosomes derived from miR-129-5p modified bone marrow mesenchymal stem cells represses ventricular remolding of mice with myocardial infarction. J Tissue Eng Regen Med 2021; 16:177-187. [PMID: 34814233 DOI: 10.1002/term.3268] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/14/2021] [Accepted: 11/15/2021] [Indexed: 12/23/2022]
Abstract
Myocardial infraction (MI) is a severe disease with great mortality. Mesenchymal stem cells-derived exosomes display protection against MI. MicroRNA-129-5p was reported to exert anti-inflammation activity by targeting high mobility group box 1 (HMGB1). In the present study, the effects of MSCs derived exosomes overexpressing miR-129-5p on MI were evaluated. Bone marrow mesenchymal stem cells (BMSCs) were transfected with miR-129-5p for exosomes isolation. Myocardial infraction mice model was established and administrated exosomes overexpressing miR-129-5p. The cardiac function, expression of HMGB1, inflammatory cytokines, apoptosis and fibrosis in heart tissues were measured. miR-129-5p inhibited HMGB1 expression in BMSCs. Myocardial infraction mice treated with exosomes overexpressing miR-129-5p had enhanced cardiac function and decreased expression of HMGB1 and production of inflammatory cytokines. Exosomes overexpressing miR-129-5p further prevented apoptosis and fibrosis. Exosome-mediated transfer of miR-129-5p suppressed inflammation in MI mice by targeting HMGB1.
Collapse
Affiliation(s)
- Shuo Wang
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Jingjie Dong
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Liu Li
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Rubing Wu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Lei Xu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Yanchun Ren
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| | - Xitian Hu
- Department of Cardiology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
33
|
Chen H, Li G, Zhang J, Zheng T, Chen Q, Zhang Y, Yang F, Wang C, Nie H, Zheng B, Gong Q. Sodium butyrate ameliorates Schistosoma japonicum-induced liver fibrosis by inhibiting HMGB1 expression. Exp Parasitol 2021; 231:108171. [PMID: 34736899 DOI: 10.1016/j.exppara.2021.108171] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/16/2021] [Accepted: 10/31/2021] [Indexed: 11/25/2022]
Abstract
Schistosomiasis is a prevalent zoonotic parasitic disease caused by schistosomes. Its main threat to human health is hepatic granuloma and fibrosis due to worm eggs. Praziquantel remains the first choice for the treatment of schistosomiasis but has limited benefit in treating liver fibrosis. Therefore, the need to develop effective drugs for treating schistosomiasis-induced hepatic fibrosis is urgent. High-mobility group box 1 protein (HMGB1) is a potential immune mediator that is highly associated with the development of some fibrotic diseases and may be involved in the liver pathology of schistosomiasis. We speculated that HMGB1 inhibitors could have an anti-fibrotic effect. Sodium butyrate (SB), a potent inhibitor of HMGB1, has shown anti-inflammatory activity in some animal disease models. In this study, we evaluated the effects of SB on a murine schistosomiasis model. Mice were percutaneously infected with 20 ± 2 cercariae of Schistosoma japonicum. SB (500 mg/kg/day) was administered every 3 days for the entire experiment period. The activity of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), liver histopathology, HMGB1 expression, and the levels of interferon gamma (IFN-γ), transforming growth factor-β1 (TGF-β1), and interleukin-6 (IL-6) in serum were analyzed. SB reduced hepatic granuloma and fibrosis of schistosomiasis, reflected by the decreased levels of ALT and AST in serum and the reduced expression of pro-inflammatory and fibrogenic cytokines (IFN-γ, TGF-β1, and IL-6). The protective effect could be attributable to the inhibition of the expression of HMGB1 and release by SB.
Collapse
Affiliation(s)
- Hui Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Gang Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Department of Gastroenterology, Jingmen Second People's Hospital, Jingmen, Hubei Province, 448000, PR China
| | - Jianqiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Ting Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Qianglin Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Yanxiang Zhang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Fei Yang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Chao Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China; Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei Province, 434023, PR China.
| |
Collapse
|
34
|
Sgrignani J, Cecchinato V, Fassi EMA, D'Agostino G, Garofalo M, Danelon G, Pedotti M, Simonelli L, Varani L, Grazioso G, Uguccioni M, Cavalli A. Systematic Development of Peptide Inhibitors Targeting the CXCL12/HMGB1 Interaction. J Med Chem 2021; 64:13439-13450. [PMID: 34510899 DOI: 10.1021/acs.jmedchem.1c00852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
During inflammatory reactions, the production and release of chemotactic factors guide the recruitment of selective leukocyte subpopulations. The alarmin HMGB1 and the chemokine CXCL12, both released in the microenvironment, can form a heterocomplex, which exclusively acts on the chemokine receptor CXCR4, enhancing cell migration, and in some pathological conditions such as rheumatoid arthritis exacerbates the immune response. An excessive cell influx at the inflammatory site can be diminished by disrupting the heterocomplex. Here, we report the computationally driven identification of the first peptide (HBP08) binding HMGB1 and selectively inhibiting the activity of the CXCL12/HMGB1 heterocomplex. Furthermore, HBP08 binds HMGB1 with the highest affinity reported so far (Kd of 0.8 ± 0.4 μM). The identification of this peptide represents an important step toward the development of innovative pharmacological tools for the treatment of severe chronic inflammatory conditions characterized by an uncontrolled immune response.
Collapse
Affiliation(s)
- Jacopo Sgrignani
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Valentina Cecchinato
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Enrico M A Fassi
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland.,Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milan, Italy
| | - Gianluca D'Agostino
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Maura Garofalo
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Gabriela Danelon
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Mattia Pedotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Luca Simonelli
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Luca Varani
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland
| | - Giovanni Grazioso
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, 20133 Milan, Italy
| | - Mariagrazia Uguccioni
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20090 Milan, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Università della Svizzera italiana, CH-6500 Bellinzona, Switzerland.,Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Zmijewska AA, Zmijewski JW, Becker EJ, Benavides GA, Darley-Usmar V, Mannon RB. Bioenergetic maladaptation and release of HMGB1 in calcineurin inhibitor-mediated nephrotoxicity. Am J Transplant 2021; 21:2964-2977. [PMID: 33724664 PMCID: PMC8429074 DOI: 10.1111/ajt.16561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 02/11/2021] [Accepted: 03/01/2021] [Indexed: 01/25/2023]
Abstract
Calcineurin inhibitors (CNIs) are potent immunosuppressive agents, universally used following solid organ transplantation to prevent rejection. Although effective, the long-term use of CNIs is associated with nephrotoxicity. The etiology of this adverse effect is complex, and effective therapeutic interventions remain to be determined. Using a combination of in vitro techniques and a mouse model of CNI-mediated nephrotoxicity, we found that the CNIs, cyclosporine A (CsA), and tacrolimus (TAC) share a similar mechanism of tubular epithelial kidney cell injury, including mitochondrial dysfunction and release of High-Mobility Group Box I (HMGB1). CNIs promote bioenergetic reprogramming due to mitochondrial dysfunction and a shift toward glycolytic metabolism. These events were accompanied by diminished cell-to-cell adhesion, loss of the epithelial cell phenotype, and release of HMGB1. Notably, Erk1/2 inhibitors effectively diminished HMGB1 release, and similar inhibitor was observed on inclusion of pan-caspase inhibitor zVAD-FMK. In vivo, while CNIs activate tissue proremodeling signaling pathways, MAPK/Erk1/2 inhibitor prevented nephrotoxicity, including diminished HMGB1 release from kidney epithelial cells and accumulation in urine. In summary, HMGB1 is an early indicator and marker of progressive nephrotoxicity induced by CNIs. We suggest that proremodeling signaling pathway and loss of mitochondrial redox/bioenergetics homeostasis are crucial therapeutic targets to ameliorate CNI-mediated nephrotoxicity.
Collapse
Affiliation(s)
- Anna A. Zmijewska
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jaroslaw W. Zmijewski
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eugene J. Becker
- Division of Pulmonary, Allergy and Critical Care, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gloria A. Benavides
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Victor Darley-Usmar
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Roslyn B. Mannon
- Division of Nephrology, University of Alabama at Birmingham, Birmingham, Alabama,Medical Service, Veterans Affairs Medical Center, Birmingham, Alabama
| |
Collapse
|
36
|
Zhu CS, Wang W, Qiang X, Chen W, Lan X, Li J, Wang H. Endogenous Regulation and Pharmacological Modulation of Sepsis-Induced HMGB1 Release and Action: An Updated Review. Cells 2021; 10:2220. [PMID: 34571869 PMCID: PMC8469563 DOI: 10.3390/cells10092220] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/13/2021] [Accepted: 08/24/2021] [Indexed: 12/12/2022] Open
Abstract
Sepsis remains a common cause of death in intensive care units, accounting for approximately 20% of total deaths worldwide. Its pathogenesis is partly attributable to dysregulated inflammatory responses to bacterial endotoxins (such as lipopolysaccharide, LPS), which stimulate innate immune cells to sequentially release early cytokines (such as tumor necrosis factor (TNF) and interferons (IFNs)) and late mediators (such as high-mobility group box 1, HMGB1). Despite difficulties in translating mechanistic insights into effective therapies, an improved understanding of the complex mechanisms underlying the pathogenesis of sepsis is still urgently needed. Here, we review recent progress in elucidating the intricate mechanisms underlying the regulation of HMGB1 release and action, and propose a few potential therapeutic candidates for future clinical investigations.
Collapse
Affiliation(s)
- Cassie Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Wei Wang
- School of Plant and Environmental Sciences, Virginia Tech, Blacksburg, VA 24061, USA;
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| | - Xiqian Lan
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
| | - Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA; (C.S.Z.); (X.Q.); (W.C.); (X.L.); (J.L.)
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY 11549, USA
| |
Collapse
|
37
|
Yang H, Zeng Q, Silverman HA, Gunasekaran M, George SJ, Devarajan A, Addorisio ME, Li J, Tsaava T, Shah V, Billiar TR, Wang H, Brines M, Andersson U, Pavlov VA, Chang EH, Chavan SS, Tracey KJ. HMGB1 released from nociceptors mediates inflammation. Proc Natl Acad Sci U S A 2021; 118:e2102034118. [PMID: 34385304 PMCID: PMC8379951 DOI: 10.1073/pnas.2102034118] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Inflammation, the body's primary defensive response system to injury and infection, is triggered by molecular signatures of microbes and tissue injury. These molecules also stimulate specialized sensory neurons, termed nociceptors. Activation of nociceptors mediates inflammation through antidromic release of neuropeptides into infected or injured tissue, producing neurogenic inflammation. Because HMGB1 is an important inflammatory mediator that is synthesized by neurons, we reasoned nociceptor release of HMGB1 might be a component of the neuroinflammatory response. In support of this possibility, we show here that transgenic nociceptors expressing channelrhodopsin-2 (ChR2) directly release HMGB1 in response to light stimulation. Additionally, HMGB1 expression in neurons was silenced by crossing synapsin-Cre (Syn-Cre) mice with floxed HMGB1 mice (HMGB1f/f). When these mice undergo sciatic nerve injury to activate neurogenic inflammation, they are protected from the development of cutaneous inflammation and allodynia as compared to wild-type controls. Syn-Cre/HMGB1fl/fl mice subjected to experimental collagen antibody-induced arthritis, a disease model in which nociceptor-dependent inflammation plays a significant pathological role, are protected from the development of allodynia and joint inflammation. Thus, nociceptor HMGB1 is required to mediate pain and inflammation during sciatic nerve injury and collagen antibody-induced arthritis.
Collapse
Affiliation(s)
- Huan Yang
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
| | - Qiong Zeng
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Harold A Silverman
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Manojkumar Gunasekaran
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Sam J George
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Alex Devarajan
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Meghan E Addorisio
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Jianhua Li
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Téa Tsaava
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Vivek Shah
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA 15213
| | - Haichao Wang
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Michael Brines
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Valentin A Pavlov
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Donald and Barbara Zucker School of Medicine at Hofstra University, Hempstead, NY 11549
| | - Eric H Chang
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Donald and Barbara Zucker School of Medicine at Hofstra University, Hempstead, NY 11549
| | - Sangeeta S Chavan
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Donald and Barbara Zucker School of Medicine at Hofstra University, Hempstead, NY 11549
| | - Kevin J Tracey
- Laboratory of Biomedical Sciences, Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030;
- The Elmezzi Graduate School of Molecular Medicine, Manhasset, NY 11030
- Donald and Barbara Zucker School of Medicine at Hofstra University, Hempstead, NY 11549
| |
Collapse
|
38
|
Devaraj A, Novotny LA, Robledo-Avila FH, Buzzo JR, Mashburn-Warren L, Jurcisek JA, Tjokro NO, Partida-Sanchez S, Bakaletz LO, Goodman SD. The extracellular innate-immune effector HMGB1 limits pathogenic bacterial biofilm proliferation. J Clin Invest 2021; 131:e140527. [PMID: 34396989 DOI: 10.1172/jci140527] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
Herein, we describe an extracellular function of the vertebrate high-mobility group box 1 protein (HMGB1) in the proliferation of bacterial biofilms. Within host cells, HMGB1 functions as a DNA architectural protein, similar to the ubiquitous DNABII family of bacterial proteins; despite that, these proteins share no amino acid sequence identity. Extracellularly, HMGB1 induces a proinflammatory immune response, whereas the DNABII proteins stabilize the extracellular DNA-dependent matrix that maintains bacterial biofilms. We showed that when both proteins converged on extracellular DNA within bacterial biofilms, HMGB1, unlike the DNABII proteins, disrupted biofilms both in vitro (including the high-priority ESKAPEE pathogens) and in vivo in 2 distinct animal models, albeit with induction of a strong inflammatory response that we attenuated by a single engineered amino acid change. We propose a model where extracellular HMGB1 balances the degree of induced inflammation and biofilm containment without excessive release of biofilm-resident bacteria.
Collapse
Affiliation(s)
- Aishwarya Devaraj
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Laura A Novotny
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Frank H Robledo-Avila
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - John R Buzzo
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Lauren Mashburn-Warren
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Joseph A Jurcisek
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Natalia O Tjokro
- Division of Periodontology, Diagnostic Sciences, and Dental Hygiene, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Santiago Partida-Sanchez
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Lauren O Bakaletz
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Steven D Goodman
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
39
|
Zhao Y, Pu M, Zhang J, Wang Y, Yan X, Yu L, He Z. Recent advancements of nanomaterial-based therapeutic strategies toward sepsis: bacterial eradication, anti-inflammation, and immunomodulation. NANOSCALE 2021; 13:10726-10747. [PMID: 34165483 DOI: 10.1039/d1nr02706a] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sepsis is a life threatening disease that is caused by a dysregulated host immune response to infection, resulting in tissue damage and organ dysfunction, which account for a high in-hospital mortality (approximately 20%). However, there are still no effective and specific therapeutics for clinical sepsis management. Nanomaterial-based strategies have emerged as promising tools for improving the therapeutic efficacy of sepsis by combating lethal bacterial infection, modulating systemic inflammatory response, preventing multiple organ failure, etc. This review has comprehensively summarized the recent advancements in nanomaterial-based strategies for the management of sepsis and severe complications, in which those nanosystems act either as inherent therapeutics or as nanocarriers for the precise delivery of agents. These formulations mechanically possess antibacterial, anti-inflammatory, immunomodulatory, and anti-oxidative effects, achieving multifunctional synergistic treatment efficacy against sepsis. Furthermore, several cell membrane-derived biomimetic nanoplatforms have been used as decoys to trap and neutralize the pathogenic toxins. The critical role of other adjuvant therapies in sepsis management, including the combination of nanotechnology and stem cell therapy, is also highlighted. Overall, this review provides insights into innovative nanotechnology-based strategies applied in sepsis treatment.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Minju Pu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Jingwen Zhang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Yanan Wang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Xuefeng Yan
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| | - Zhiyu He
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, China.
| |
Collapse
|
40
|
Zhu ZH, Li X, He LF, Cai HF, Ye B, Wu ZM. Glycyrrhizic acid, as an inhibitor of HMGB1, alleviates bleomycin-induced pulmonary toxicity in mice through the MAPK and Smad3 pathways. Immunopharmacol Immunotoxicol 2021; 43:461-470. [PMID: 34142927 DOI: 10.1080/08923973.2021.1939371] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
AIM High-mobility group box 1 (HMGB1) protein has been noticed particularly for its pivotal role in several pathologies. However, the relevance between HMGB1 and pathological progress in lung toxicity still remains unclear. In the study, we evaluated the effect of glycyrrhizic acid as an HMGB1 inhibitor on the early inflammation and late fibrosis in bleomycin-induced pulmonary toxicity in mice. METHODS We established a bleomycin-induced pulmonary toxicity model to detect the relevance between HMGB1 and pathological changes in the early inflammatory and late fibrotic stages. RESULTS We found that bleomycin-induced increase in inflammatory cytokines interleukin (IL)-β1, tumor necrosis factor (TNF)-α, monocyte chemotactic protein (MCP)-1, and inflammatory lesions in lung tissue in the early stage of the model. However, markers of fibrosis such as transforming growth factor (TGF)-β1 and α-smooth muscle actin (α-SMA) were significantly elevated on day 7 after bleomycin instillation. Interestingly, HMGB1 also began to rise on day 7, rather than in the early inflammatory phase. However, early (from day 0 to 14 after bleomycin instillation) or late (from day 14 to 28) intervention with HMGB1 neutralizing antibody or glycyrrhizic acid alleviated inflammation and fibrosis through down-regulating the inflammatory signaling mitogen-activated protein kinase (MAPK) and fibrotic signaling Smad3 pathway. CONCLUSION Our results suggested that HMGB1 mediates both inflammation and fibrosis in this model. The development of high-potency and low-toxicity HMGB1 inhibitors may be a class of potential drugs for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhen-Hua Zhu
- Taizhou Central Hospital, School of Medicine of Taizhou University, Taizhou, China
| | - Xing Li
- Taizhou Central Hospital, School of Medicine of Taizhou University, Taizhou, China
| | - Lin-Feng He
- Taizhou Central Hospital, School of Medicine of Taizhou University, Taizhou, China
| | - He-Fei Cai
- Taizhou Central Hospital, School of Medicine of Taizhou University, Taizhou, China
| | - Bin Ye
- Taizhou Central Hospital, School of Medicine of Taizhou University, Taizhou, China
| | - Zhong-Min Wu
- Department of Basic Medical Sciences, School of Medicine of Taizhou University, Taizhou, China
| |
Collapse
|
41
|
Chen W, Qiang X, Wang Y, Zhu S, Li J, Babaev A, Yang H, Gong J, Becker L, Wang P, Tracey KJ, Wang H. Identification of tetranectin-targeting monoclonal antibodies to treat potentially lethal sepsis. Sci Transl Med 2021; 12:12/539/eaaz3833. [PMID: 32295901 DOI: 10.1126/scitranslmed.aaz3833] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 03/09/2020] [Indexed: 12/12/2022]
Abstract
For the clinical management of sepsis, antibody-based strategies have only been attempted to antagonize proinflammatory cytokines but not yet been tried to target harmless proteins that may interact with these pathogenic mediators. Here, we report an antibody strategy to intervene in the harmful interaction between tetranectin (TN) and a late-acting sepsis mediator, high-mobility group box 1 (HMGB1), in preclinical settings. We found that TN could bind HMGB1 to reciprocally enhance their endocytosis, thereby inducing macrophage pyroptosis and consequent release of lactate dehydrogenase and apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain. The genetic depletion of TN expression or supplementation of exogenous TN protein at subphysiological doses distinctly affected the outcomes of potentially lethal sepsis, revealing a previously underappreciated beneficial role of TN in sepsis. Furthermore, the administration of domain-specific polyclonal and monoclonal antibodies effectively inhibited TN/HMGB1 interaction and endocytosis and attenuated the sepsis-induced TN depletion and tissue injury, thereby rescuing animals from lethal sepsis. Our findings point to a possibility of developing antibody strategies to prevent harmful interactions between harmless proteins and pathogenic mediators of human diseases.
Collapse
Affiliation(s)
- Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Yongjun Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Shu Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Ariella Babaev
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA
| | - Huan Yang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Jonathan Gong
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Lance Becker
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Ping Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Kevin J Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, 350 Community Drive, Manhasset, NY 11030, USA. .,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd., Hempstead, NY 11549, USA
| |
Collapse
|
42
|
The Effect and Regulatory Mechanism of High Mobility Group Box-1 Protein on Immune Cells in Inflammatory Diseases. Cells 2021; 10:cells10051044. [PMID: 33925132 PMCID: PMC8145631 DOI: 10.3390/cells10051044] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
High mobility group box-1 protein (HMGB1), a member of the high mobility group protein superfamily, is an abundant and ubiquitously expressed nuclear protein. Intracellular HMGB1 is released by immune and necrotic cells and secreted HMGB1 activates a range of immune cells, contributing to the excessive release of inflammatory cytokines and promoting processes such as cell migration and adhesion. Moreover, HMGB1 is a typical damage-associated molecular pattern molecule that participates in various inflammatory and immune responses. In these ways, it plays a critical role in the pathophysiology of inflammatory diseases. Herein, we review the effects of HMGB1 on various immune cell types and describe the molecular mechanisms by which it contributes to the development of inflammatory disorders. Finally, we address the therapeutic potential of targeting HMGB1.
Collapse
|
43
|
Local administration of HMGB-1 promotes bone regeneration on the critical-sized mandibular defects in rabbits. Sci Rep 2021; 11:8950. [PMID: 33903607 PMCID: PMC8076241 DOI: 10.1038/s41598-021-88195-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 04/08/2021] [Indexed: 01/02/2023] Open
Abstract
Reconstruction of a critical-sized osseous defect is challenging in maxillofacial surgery. Despite novel treatments and advances in supportive therapies, severe complications including infection, nonunion, and malunion can still occur. Here, we aimed to assess the use of a beta-tricalcium phosphate (β-TCP) scaffold loaded with high mobility group box-1 protein (HMGB-1) as a novel critical-sized bone defect treatment in rabbits. The study was performed on 15 specific pathogen-free New Zealand rabbits divided into three groups: Group A had an osseous defect filled with a β-TCP scaffold loaded with phosphate-buffered saline (PBS) (100 µL/scaffold), the defect in group B was filled with recombinant human bone morphogenetic protein 2 (rhBMP-2) (10 µg/100 µL), and the defect in group C was loaded with HMGB-1 (10 µg/100 µL). Micro-computed tomography (CT) examination demonstrated that group C (HMGB-1) showed the highest new bone volume ratio, with a mean value of 66.5%, followed by the group B (rhBMP-2) (31.0%), and group A (Control) (7.1%). Histological examination of the HMGB-1 treated group showed a vast area covered by lamellar and woven bone surrounding the β-TCP granule remnants. These results suggest that HMGB-1 could be an effective alternative molecule for bone regeneration in critical-sized mandibular bone defects.
Collapse
|
44
|
Xue L, Guo W, Li L, Ou S, Zhu T, Cai L, Ding W, Wu W. Metabolomic profiling identifies a novel mechanism for heat stroke‑related acute kidney injury. Mol Med Rep 2021; 23:241. [PMID: 33655337 PMCID: PMC7893796 DOI: 10.3892/mmr.2021.11880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 08/20/2020] [Indexed: 11/27/2022] Open
Abstract
Heat stroke can induce a systemic inflammatory response, which may lead to multi‑organ dysfunction including acute kidney injury (AKI) and electrolyte disturbances. To investigate the pathogenesis of heat stroke (HS)‑related AKI, a mouse model of HS was induced by increasing the animal's core temperature to 41˚C. Blood samples obtained from the tail vein were used to measure plasma glucose and creatinine levels. Micro‑positron emission tomography‑computed tomography (micro‑PET/CT), H&E staining and transmission electron microscopy were conducted to examine metabolic and morphological changes in the mouse kidneys. Immunohistochemistry (IHC) and western blot analyses were performed to investigate the expression of apoptosis‑inducing factor mitochondria‑associated 2 (Aifm2), high‑mobility group box 1 (HMGB1) and receptor for advanced glycosylation end products (RAGE). Liquid chromatography‑mass spectrometry analysis was conducted to find differential metabolites and signaling pathways. The HS mouse model was built successfully, with significantly increased creatinine levels detected in the serum of HS mice compared with controls, whereas micro‑PET/CT revealed active metabolism in the whole body of HS mice. H&E and TUNEL staining revealed that the kidneys of HS mice exhibited signs of hemorrhage and apoptosis. IHC and western blotting demonstrated significant upregulation of Aifm2, HMGB1 and RAGE in response to HS. Finally, 136 differential metabolites were screened out, and enrichment of the 'biosynthesis of unsaturated fatty acids' pathway was detected. HS‑associated AKI is the renal manifestation of systemic inflammatory response syndrome, and may be triggered by the HMGB1/RAGE pathway. Metabolomics indicated increased adrenic acid, docosahexaenoic acid and eicosapentaenoic acid may serve as metabolic biomarkers for AKI in HS. The findings suggested that a correlation between the HMGB1/RAGE pathway and biosynthesis of unsaturated fatty acids may contribute to the progression of HS‑related AKI.
Collapse
Affiliation(s)
- Ling Xue
- Department of Urology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Guo
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Li
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Santao Ou
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tingting Zhu
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Liang Cai
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenfei Ding
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Weihua Wu
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
45
|
Ludes PO, de Roquetaillade C, Chousterman BG, Pottecher J, Mebazaa A. Role of Damage-Associated Molecular Patterns in Septic Acute Kidney Injury, From Injury to Recovery. Front Immunol 2021; 12:606622. [PMID: 33732235 PMCID: PMC7957065 DOI: 10.3389/fimmu.2021.606622] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Damage-associated molecular patterns (DAMPs) are a group of immunostimulatory molecules, which take part in inflammatory response after tissue injury. Kidney-specific DAMPs include Tamm-Horsfall glycoprotein, crystals, and uromodulin, released by tubular damage for example. Non-kidney-specific DAMPs include intracellular particles such as nucleus [histones, high-mobility group box 1 protein (HMGB1)] and cytosol parts. DAMPs trigger innate immunity by activating the NRLP3 inflammasome, G-protein coupled class receptors or the Toll-like receptor. Tubular necrosis leads to acute kidney injury (AKI) in either septic, ischemic or toxic conditions. Tubular necrosis releases DAMPs such as histones and HMGB1 and increases vascular permeability, which perpetuates shock and hypoperfusion via Toll Like Receptors. In acute tubular necrosis, intracellular abundance of NADPH may explain a chain reaction where necrosis spreads from cell to cell. The nature AKI in intensive care units does not have preclinical models that meet a variation of blood perfusion or a variation of glomerular filtration within hours before catecholamine infusion. However, the dampening of several DAMPs in AKI could provide organ protection. Research should be focused on the numerous pathophysiological pathways to identify the relative contribution to renal dysfunction. The therapeutic perspectives could be strategies to suppress side effect of DAMPs and to promote renal function regeneration.
Collapse
Affiliation(s)
- Pierre-Olivier Ludes
- Department of Anesthesiology and Intensive Care, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,EA 3072, Mitochondrie Stress Oxydant et Protection Musculaire, Faculté de Médecine, FRU 6702, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Charles de Roquetaillade
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| | - Benjamin Glenn Chousterman
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| | - Julien Pottecher
- Department of Anesthesiology and Intensive Care, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France.,EA 3072, Mitochondrie Stress Oxydant et Protection Musculaire, Faculté de Médecine, FRU 6702, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Alexandre Mebazaa
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, DMU Parabol, APHP.Nord, Paris, France.,Inserm U942 MASCOT, Université de Paris, Paris, France
| |
Collapse
|
46
|
Liu N, Wu Y, Wen X, Li P, Lu F, Shang H. Chronic stress promotes acute myeloid leukemia progression through HMGB1/NLRP3/IL-1β signaling pathway. J Mol Med (Berl) 2021; 99:403-414. [PMID: 33409553 DOI: 10.1007/s00109-020-02011-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/08/2020] [Accepted: 11/18/2020] [Indexed: 12/18/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy with poor prognosis and overall survival. Clinical investigations show that chronic stress is commonly present in the course of AML and associated with adverse outcome. However, the underlying molecular mechanisms are elusive. In the present study, a chronic restraint stress mouse model was established to evaluate the effect of stress on AML. We found that mice under chronic stress exhibited significantly increased liver and spleen infiltration of leukemic cells and poorer overall survival. This was accompanied by elevated cellular NLR family pyrin domain containing 3 (NLRP3) and interleukin-1β (IL-1β) in the liver or bone marrow, and secreted IL-1β in the plasma, indicating the activation of inflammasomes under chronic restraint stress. High mobility group box 1 (HMGB1) expression was markedly increased in newly diagnosed AML patients, but reduced in complete remission AML patients. The expression level of HMGB1 was positively correlated with NLRP3 mRNA in AML patients. Knockdown of HMGB1 significantly decreased NLRP3 and IL-1β expression in AML cell lines, and secreted IL-1β in supernatant of AML cell culture, while HMGB1 stimulation caused contrary effects. These results implied that HMGB1 could be involved in the regulation of inflammasome activation in AML development. Mice model showed that chronic restraint stress-facilitated proliferation and infiltration of AML cells were largely abrogated by knocking down HMGB1. Knockdown of HMGB1 also ameliorated overall survival and remarkably neutralized NLRP3 and IL-1β expression under chronic restraint stress. These findings provide evidences that chronic stress promotes AML progression via HMGB1/NLRP3/IL-1β dependent mechanism, suggesting that HMGB1 is a potential therapeutic target for AML. KEY MESSAGES: • Chronic restraint stress promoted acute myeloid leukemia (AML) progression and mediated NLRP3 inflammasome activation in xenograft mice. • HMGB1 mediated NLRP3 inflammasome activation in AML cells. • Knockdown of HMGB1 inhibited AML progression under chronic stress in vivo.
Collapse
MESH Headings
- Animals
- Bone Marrow/metabolism
- Cell Line, Tumor
- Chronic Disease
- Disease Progression
- Female
- Gene Expression Regulation, Leukemic
- Gene Knockdown Techniques
- HMGB1 Protein/antagonists & inhibitors
- HMGB1 Protein/biosynthesis
- HMGB1 Protein/genetics
- HMGB1 Protein/physiology
- Heterografts
- Humans
- Inflammasomes/metabolism
- Inflammation
- Interleukin-1beta/biosynthesis
- Interleukin-1beta/genetics
- Interleukin-1beta/physiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/physiopathology
- Liver/metabolism
- Liver/pathology
- Mice, Inbred C57BL
- NLR Family, Pyrin Domain-Containing 3 Protein/biosynthesis
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/physiology
- Neoplasm Proteins/physiology
- RNA Interference
- Remission Induction
- Restraint, Physical
- Signal Transduction/physiology
- Spleen/metabolism
- Spleen/pathology
- Stress, Physiological
- Toll-Like Receptor 4/physiology
- Mice
Collapse
Affiliation(s)
- Na Liu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Yifan Wu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
- Xinhua Hospital Chongming Branch, Shanghai Jiaotong University School of Medicine, Shanghai, 202150, China
| | - Xin Wen
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Peng Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fei Lu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Hong Shang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong, China.
| |
Collapse
|
47
|
Miao J, Zhong J, Lan J, Ye S, Ye P, Li S, You A, Chen X, Liu X, Li H. Paeonol attenuates inflammation by confining HMGB1 to the nucleus. J Cell Mol Med 2021; 25:2885-2899. [PMID: 33534963 PMCID: PMC7957162 DOI: 10.1111/jcmm.16319] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/22/2020] [Accepted: 01/09/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammation is a biological process that exists in a large number of diseases. If the magnitude or duration of inflammation becomes uncontrolled, inflammation may cause pathological damage to the host. HMGB1 and NF-κB have been shown to play pivotal roles in inflammation-related diseases. New drugs aimed at inhibiting HMGB1 expression have become a key research focus. In the present study, we showed that paeonol (Pae), the main active component of Paeonia suffruticosa, decreases the expression of inflammatory cytokines and inhibits the translocation of HMGB1 induced by lipopolysaccharide (LPS). By constructing HMGB1-overexpressing (HMGB1+ ) and HMGB1-mutant (HMGB1m ) RAW264.7 cells, we found that the nuclear HMGB1 could induce an LPS-tolerant state in RAW264.7 cells and that paeonol had no influence on the expression of inflammatory cytokines in HMGB1m RAW264.7 cells. In addition, the anti-inflammatory property of paeonol was lost in HMGB1 conditional knockout mice, indicating that HMGB1 is a target of paeonol and a mediator through which paeonol exerts its anti-inflammatory function. Additionally, we also found that HMGB1 and P50 competitively bound with P65, thus inactivating the NF-κB pathway. Our research confirmed the anti-inflammation property of paeonol and suggests that inhibiting the translocation of HMGB1 could be a new strategy for treating inflammation.
Collapse
Affiliation(s)
- Jifei Miao
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China.,Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Jun Zhong
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Jiao Lan
- Shenzhen Bao'an Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Sen Ye
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Peng Ye
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Siyan Li
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Aijia You
- School of Nursing, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianjie Chen
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Liu
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| | - Hui Li
- Research Center of Integrative Medicine, School Basic Medical Sciences, University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
48
|
Zhao H, Gu Y, Chen H. Propofol ameliorates endotoxin‑induced myocardial cell injury by inhibiting inflammation and apoptosis via the PPARγ/HMGB1/NLRP3 axis. Mol Med Rep 2021; 23:176. [PMID: 33398367 PMCID: PMC7821353 DOI: 10.3892/mmr.2020.11815] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 11/11/2020] [Indexed: 11/05/2022] Open
Abstract
Endotoxin lipopolysaccharide (LPS) is one of the primary causes of myocardial injury. Propofol confers protective effects against LPS‑induced myocardial damage; however, the biological functions and mechanisms underlying propofol are not completely understood. The present study aimed to investigate the effects of propofol on LPS‑induced myocardial injury. Primary neonatal rat cardiomyocytes were treated with LPS to establish a myocardial injury model. LDH release in the culture media was measured using a LDH assay kit. The interactions between NLR family pyrin domain containing 3 (NLRP3), apoptosis‑associated speck‑like protein containing A CARD (ASC) and pro‑caspase‑1 were determined using a co‑immunoprecipitation assay. Cell viability was measured using an MTT assay, and the levels of cell apoptosis were determined using flow cytometry, JC‑1 staining (mitochondrial membrane potential) and caspase‑3 activity assays. The mRNA expression levels of TNF‑α, IL‑6, IL‑1β and IL‑18, and the protein expression levels of NLRP3, ASC, pro‑caspase‑1, caspase‑1 p10, pro‑IL‑1β, IL‑1β, pro‑IL‑18, IL‑18, high mobility group box‑1 (HMGB1) and peroxisome proliferator‑activated receptor γ (PPARγ) were analyzed using reverse transcription‑quantitative PCR and western blotting analyses, respectively. ELISAs were performed to measure the production of inflammatory mediators, including TNF‑α, IL‑6, IL‑1β and IL‑18. The present results demonstrated that pretreatment with propofol significantly attenuated LPS‑induced neonatal rat cardiomyocyte injury in a concentration‑ and time‑dependent manner. Propofol pretreatment also significantly inhibited LPS‑induced cardiomyocyte inflammation and apoptosis. The results suggested that propofol pretreatment inactivated HMGB1‑dependent NLRP3 inflammasome signaling, which involved PPARγ activation. Therefore, the results indicated that propofol reduced endotoxin‑induced cardiomyocyte injury by inhibiting inflammation and apoptosis via the PPARγ/HMGB1/NLRP3 axis, suggesting that propofol may serve as a potential therapeutic agent for septic myocardial damage.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ying Gu
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hai Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
49
|
Feng E, Wang J, Wang X, Wang Z, Chen X, Zhu X, Hou W. Inhibition of HMGB1 Might Enhance the Protective Effect of Taxifolin in Cardiomyocytes via PI3K/AKT Signaling Pathway. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2021; 20:316-332. [PMID: 34567165 PMCID: PMC8457741 DOI: 10.22037/ijpr.2020.113584.14384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Cardiovascular diseases (CVD) affect millions of people and spend a lot of medical costs around the world each year. Taxifolin is a natural anti-oxidative reagent obtained from multiple plants and exhibits a wide range of pharmacological effects. High mobility group box protein 1 (HMGB1) is expressed in multiple types of cells in the extracellular environment, regulating the pro-inflammatory process. Here, we detected the viability of cells using MTT assay, and the expression of each target protein was detected using western blotting analysis. The expression of each target mRNA was detected using the qPCR method, and the concentration of each cytokine in serum samples was detected using the ELISA method. In this study, we found that taxifolin could decrease the expression of hypoxia-inducible factor-1α (HIF-1α) while increasing the expression of endothelial nitric oxide synthase (eNOS), presented a protective role. Besides, taxifolin could also increase the expression of vascular endothelial growth factor-α (VEGF-α), transforming growth factor-β (TGF-β) and fibroblast growth factor21 (FGF21), resulting in viability rate increasing. And these effects were mediated by phosphatidylinositol 3-hydroxy kinase (PI3K)/AKT/mTOR signaling pathway; a similar trend was also observed in HMGB1 knockdown mice. We also found that inhibition of HMGB1 could enhance the cardioprotective effect of taxifolin and might be a new therapeutic strategy for cardiovascular disease.
Collapse
Affiliation(s)
- Erjun Feng
- Department of Cardiology, Fourth Center Hospital of Tianjin, Tianjin, China, 300000.
- E. F. and J. W. and X. W. contributed equally to this work.
| | - Jian Wang
- Department of Cardiology, Fourth Center Hospital of Tianjin, Tianjin, China, 300000.
- E. F. and J. W. and X. W. contributed equally to this work.
| | - Xinwei Wang
- Oncology Department of Characteristic Medical Center of PAF, Tianjin, China, 300162.
- E. F. and J. W. and X. W. contributed equally to this work.
| | - Zhenguo Wang
- Medical Research Department of Characteristic Medical Center of PAF, Tianjin, China, 300162.
| | - Xiaochu Chen
- Medical Research Department of Characteristic Medical Center of PAF, Tianjin, China, 300162.
| | - Xu Zhu
- Second Department of Neurology, Central Hospital of Handan, Handan, China, 056000.
| | - Wenli Hou
- Cadre Ward of Characteristic Medical Center of PAF, Tianjin, China, 300162.
| |
Collapse
|
50
|
Elmehy DA, Ismail HI, Soliman NA, Amer BS, Elkaliny HH, El-Ebiary AA, Gamea GA. Oxidative stress mediated apoptotic potential of mefloquine on experimental trichinellosis. Acta Trop 2021; 213:105760. [PMID: 33221280 DOI: 10.1016/j.actatropica.2020.105760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 11/02/2020] [Accepted: 11/12/2020] [Indexed: 10/23/2022]
Abstract
Conventional anthelmintics such as albendazole could not achieve complete cure of trichinellosis till now. The antimalarial mefloquine mediates oxidative stress and disrupts lysosomal functions leading to cell death. Therefore, the aim of this work was to investigate the effect of mefloquine on experimental acute and chronic trichinellosis and to clarify the possible mechanisms of such effects. Mice were divided into four groups; Group I: Uninfected untreated control (20 mice); Group II: Infected untreated control (40 mice); Group III: infected and treated with albendazole (400 mg/kg) (40 mice); Group IV: infected and treated with mefloquine (300 mg/kg) (40 mice). All infected treated groups were equally subdivided into 2 subgroups; (a) treated on the 2nd day post infection (dpi) for 3 days, (b) treated on the 35th dpi for 5 days. Parasitological adults and larvae counting besides immunohistopathological examination of intestines and muscles were done. Biochemical assay of oxidant/antioxidant status, apoptotic, cytoprotective and inflammatory biomarkers in intestinal and muscle homogenates were achieved. Results showed that both albendazole and mefloquine significantly reduced adults and larvae counts with higher efficacy of albendazole in the intestinal phase and superiority of mefloquine in the muscle phase. The superiority of mefloquine was indicated by increased inflammatory immune infiltration and decreased anti-apoptotic immunohistochemical markers expression in both jejunal and muscle tissues. Biochemically, mefloquine treatment showed highly significant oxidative, apoptotic and inflammatory effects. So, our results suggest that mefloquine might be a superior treatment for chronic trichinellosis.
Collapse
|