1
|
Li H, Peng MX, Yang RX, Chen JX, Wang YM, Wang PX, Hu YH, Pan DY, Liu PQ, Lu J. SNX3 mediates heart failure by interacting with HMGB1 and subsequently facilitating its nuclear-cytoplasmic translocation. Acta Pharmacol Sin 2025; 46:964-975. [PMID: 39753981 PMCID: PMC11950316 DOI: 10.1038/s41401-024-01436-z] [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: 07/30/2023] [Accepted: 11/17/2024] [Indexed: 03/17/2025]
Abstract
Sorting nexins (SNXs) as the key regulators of sorting cargo proteins are involved in diverse diseases. SNXs can form the specific reverse vesicle transport complex (SNXs-retromer) with vacuolar protein sortings (VPSs) to sort and modulate recovery and degradation of cargo proteins. Our previous study has shown that SNX3-retromer promotes both STAT3 activation and nuclear translocation in cardiomyocytes, suggesting that SNX3 might be a critical regulator in the heart. In this study we investigated the role of SNX3 in the development of pathological cardiac hypertrophy and heart failure. We generated abdominal aortic constriction (AAC) rat model and transverse aortic constriction (TAC) mouse model; hypertrophic neonatal rat cardiomyocytes (NRCMs) were induced by exposure to isoproterenol (10 μM). We showed that the expression of SNX3 was significantly upregulated in ISO-treated NRCMs and in the failing heart of AAC rats. Overexpression of SNX3 by intramyocardial injection of Ad-SNX3 induced heart failure in rats, and increased the susceptibility of NRCMs to ISO-induced myocardial injury in vitro. In contrast, conditional knockout of SNX3 in cardiac tissue in mice rescued the detrimental heart function in TAC mice, and knockdown of SNX3 protected against ISO-induced injury in NRCMs and AAC rats. We then conducted immunoprecipitation-based mass spectrometry and localized surface plasmon resonance, and demonstrated a direct interaction between SNX3-retromer and high mobility group box 1 (HMGB1), which mediated the efflux of nuclear HMGB1. Moreover, overexpression of HMGB1 in NRCMs inhibited the pro-hypertrophic effects of SNX3, whereas knockdown of HMGB1 abolished the protective effect of SNX3-deficiency. These results suggest that HMGB1 might be a direct cargo protein of SNX3-retromer, and its interaction with SNX3 promotes its efflux from the nucleus, leading to the pathological development of heart failure.
Collapse
Affiliation(s)
- Hong Li
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- The Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ming-Xia Peng
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Rui-Xue Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- The Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jian-Xing Chen
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yue-Mei Wang
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Pan-Xia Wang
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yue-Huai Hu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Di-Yi Pan
- Department of Pharmacy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510378, China
| | - Pei-Qing Liu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Yang Y, Wu A, Deng AN, Liu H, Lan Q, Mazhar M, Xue JY, Chen MT, Luo G, Liu MN. Macrophages after myocardial infarction: Mechanisms for repairing and potential as therapeutic approaches. Int Immunopharmacol 2024; 143:113562. [PMID: 39536484 DOI: 10.1016/j.intimp.2024.113562] [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: 09/21/2024] [Revised: 10/20/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Macrophages - one of the crucial immune cells, are recruited to the cardiac tissue by chemokines, cytokines and upregulated endothelial adhesion molecules after myocardial infarction (MI). During the course of inflammation in the cardiac tissue, necrotic cells and matrix debris is phagocytosed by M1 macrophages. During the resolution phase of cardiac inflammation, M2 macrophages promote cardiac recovery. Suppression or over expression of both the M1 and M2 macrophage subtypes significantly affect the reparation of infarction. Stem cells therapy, cytokine regulation and immune cells therapy are considered as effective interventions to regulate the phenotypic transformation of cardiac macrophages after MI. Intervention with macrophages in the myocardium has shown unique advantages. In this review, the mechanisms and role of macrophages in the development of MI are elaborated in detail, the promising therapeutic methods for regulating macrophage phenotypes, their limitations and possible future research directions are discussed.
Collapse
Affiliation(s)
- You Yang
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ai Wu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - An-Ni Deng
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Hao Liu
- Department of Pediatrics, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Qi Lan
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Maryam Mazhar
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Jin-Yi Xue
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Ming-Tai Chen
- Department of Cardiovascular Disease, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China.
| | - Gang Luo
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| | - Meng-Nan Liu
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
3
|
Ruggieri E, Di Domenico E, Locatelli AG, Isopo F, Damanti S, De Lorenzo R, Milan E, Musco G, Rovere-Querini P, Cenci S, Vénéreau E. HMGB1, an evolving pleiotropic protein critical for cellular and tissue homeostasis: Role in aging and age-related diseases. Ageing Res Rev 2024; 102:102550. [PMID: 39427887 DOI: 10.1016/j.arr.2024.102550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/05/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
Aging is a universal biological process characterized by a progressive, cumulative decline in homeostatic capabilities and physiological functions, which inevitably increases vulnerability to diseases. A number of molecular pathomechanisms and hallmarks of aging have been recognized, yet we miss a thorough understanding of their complex interconnectedness. This review explores the molecular and cellular mechanisms underlying human aging, with a focus on the multiple roles of high mobility group Box 1 protein (HMGB1), the archetypal damage-associated molecular pattern (DAMP) molecule. In the nucleus, this non-histone chromatin-associated protein functions as a DNA chaperone and regulator of gene transcription, influencing DNA structure and gene expression. Moreover, this versatile protein can translocate to the cytoplasm to orchestrate other processes, such as autophagy, or be unconventionally secreted into the extracellular environment, where it acts as a DAMP, combining inflammatory and regenerative properties. Notably, lower expression of HMGB1 within the cell and its heightened extracellular release have been associated with diverse age-associated traits, making it a suitable candidate as a universal biomarker of aging. In this review, we outline the evidence implicating HMGB1 in aging, also in light of an evolutionary perspective on its functional pleiotropy, and propose critical issues that need to be addressed to gauge the value of HMGB1 as a potential biomarker across age-related diseases and therapeutic target to promote healthy longevity.
Collapse
Affiliation(s)
- Elena Ruggieri
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Erika Di Domenico
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Flavio Isopo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Sarah Damanti
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Rebecca De Lorenzo
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Enrico Milan
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | | | - Patrizia Rovere-Querini
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| | - Simone Cenci
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| | - Emilie Vénéreau
- IRCCS Ospedale San Raffaele, Milano, Italy; Università Vita-Salute San Raffaele, Milano, Italy.
| |
Collapse
|
4
|
Zhong Q, Zhao B, She X, Liu X. HMGA2 as a prognostic and immune biomarker in hepatocellular carcinoma: Comprehensive analysis of the HMG family and experiments validation. PLoS One 2024; 19:e0311204. [PMID: 39591457 PMCID: PMC11594397 DOI: 10.1371/journal.pone.0311204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/15/2024] [Indexed: 11/28/2024] Open
Abstract
The molecular mechanisms underlying hepatocellular carcinoma (HCC) are complex and not fully understood. This study aims to explore the expression and clinical significance of High Mobility Group (HMG) proteins in HCC to identify potential prognostic biomarkers and therapeutic targets. Bioinformatic analyses were performed using data from The Cancer Genome Atlas (TCGA) and other databases. Expression levels of HMGs were validated in HCC cell lines using qRT-PCR, and functional studies were conducted by knocking down HMGA2.HMG family members, particularly HMGA1, HMGA2, HMGB2, and HMGN1, were significantly upregulated in HCC tissues compared to normal tissues. High expression levels of these proteins were associated with poor overall survival and disease-specific survival in HCC patients. Knockdown of HMGA2 in HCC cell lines led to reduced cell proliferation, migration, and invasion. HMGA2, along with other HMG family members, emerges as a potential prognostic biomarker and therapeutic target in HCC. This study provides new insights into the role of HMG proteins in HCC progression.
Collapse
Affiliation(s)
- Qiangqiang Zhong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Laboratory of Metabolic Abnormalities and Vascular Aging Huazhong University of Science and Technology, Wuhan, PR China
| | - Baokang Zhao
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| | - Xiao She
- Department of Gastroenterology, Xi’an Jiaotong University Second Affiliated Hospital, Xi’an, PR China
| | - Xiangjie Liu
- Department of Geriatrics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, PR China
| |
Collapse
|
5
|
Bi C, Wang D, Hao B, Yang T. Snhg14/miR-181a-5p axis-mediated "M1" macrophages aggravate LPS-induced myocardial cell injury. Heliyon 2024; 10:e37104. [PMID: 39309894 PMCID: PMC11414504 DOI: 10.1016/j.heliyon.2024.e37104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
An increasing number of studies have suggested that macrophages participate in sepsis-induced myocardial injury. Our study highlights the function and mechanism of the lncRNA Snhg14 in "M1" polarized macrophage-mediated myocardial cell damage. Lipopolysaccharide (LPS) was used to treat H9c2 cells to construct an in vitro myocardial injury model. M1 and M2 polarization of RAW264.7 cells were induced and the exosomes were obtained from the supernatant through ultracentrifugation. Moreover, cecal ligation and puncture (CLP) surgery was implemented to establish a mouse sepsis-induced myocardial injury model, and Snhg14 was knocked down with sh-Snhg14. The results showed that the conditioned medium (CM) and the exosomes (Exo) of M1 macrophages substantially augmented LPS-induced apoptosis and oxidative stress in myocardial cells. Notably, M1-CM and M1-Exo contributed to nearly 50 % of myocardial cell viability decline. Snhg14 was highly expressed in M1 macrophages and exosomes derived from M1-MΦ (M1-Exo). Snhg14 overexpression aggravated myocardial cell damage and increased 10 to 50 times expression of proinflammatory cytokines in MΦ. Snhg14 knockdown reversed M1-Exo-mediated myocardial cell damage and inhibited the production of proinflammatory cytokines (50 %-75 % decline) of MΦ. Moreover, Snhg14 targeted and inhibited miR-181a-5p expression. miR-181a-5p upregulation partly reversed Snhg4 overexpression-mediated myocardial cell damage and MΦ activation. In vivo, sh-Snhg14 dramatically ameliorated cardiac damage in septic mice by enhancing miR-181a-5p and inhibiting the HMGB1/NF-κB pathway. In conclusion, "M1" macrophage-derived exosomal Snhg14 aggravates myocardial cell damage by modulating the miR-181a-5p/HMGB1/NF-κB pathway.
Collapse
Affiliation(s)
- Chenglong Bi
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Dejin Wang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| | - Bin Hao
- Cardiovascular Surgery, Shandong University Zibo Central Hospital, Zibo, 255000, China
| | - Tianxiao Yang
- Department of Cardiology, Shandong University Zibo Central Hospital, Zibo, 255000, Shandong, China
| |
Collapse
|
6
|
Slick RA, Sutton J, Haberman M, O'Brien BS, Tinklenberg JA, Mardikar A, Prom MJ, Beatka M, Gartz M, Vanden Avond MA, Siebers E, Mack DL, Gonzalez JP, Ebert AD, Nagaraju K, Lawlor MW. High mobility group box 1 (HMGB1) is a potential disease biomarker in cell and mouse models of Duchenne muscular dystrophy. Biol Open 2024; 13:bio060542. [PMID: 39158383 PMCID: PMC11391821 DOI: 10.1242/bio.060542] [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: 05/26/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disorder affecting 1:3500 male births and is associated with myofiber degeneration, regeneration, and inflammation. Glucocorticoid treatments have been the standard of care due to immunomodulatory/immunosuppressive properties but novel genetic approaches, including exon skipping and gene replacement therapy, are currently being developed. The identification of additional biomarkers to assess DMD-related inflammatory responses and the potential efficacy of these therapeutic approaches are thus of critical importance. The current study uses RNA sequencing of skeletal muscle from two mdx mouse models to identify high mobility group box 1 (HMGB1) as a candidate biomarker potentially contributing to DMD-related inflammation. HMGB1 protein content was increased in a human iPSC-derived skeletal myocyte model of DMD and microdystrophin treatment decreased HMGB1 back to control levels. In vivo, HMGB1 protein levels were increased in vehicle treated B10-mdx skeletal muscle compared to B10-WT and significantly decreased in B10-mdx animals treated with adeno-associated virus (AAV)-microdystrophin. However, HMGB1 protein levels were not increased in D2-mdx skeletal muscle compared to D2-WT, demonstrating a strain-specific difference in DMD-related immunopathology.
Collapse
Affiliation(s)
- Rebecca A. Slick
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jessica Sutton
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Margaret Haberman
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Benjamin S. O'Brien
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer A. Tinklenberg
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aashay Mardikar
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mariah J. Prom
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Margaret Beatka
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| | - Melanie Gartz
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mark A. Vanden Avond
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Emily Siebers
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - David L. Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA 98104, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98104, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98104, USA
| | | | - Allison D. Ebert
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kanneboyina Nagaraju
- AGADA BioSciences Inc., Halifax, Nova Scotia, B3H0A8, Canada
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, SUNY. Binghamton, NY 13902, USA
| | - Michael W. Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Diverge Translational Science Laboratory, Milwaukee, WI 53204, USA
| |
Collapse
|
7
|
Tian H, Liu Q, Yu X, Cao Y, Huang X. Damage-associated molecular patterns in viral infection: potential therapeutic targets. Crit Rev Microbiol 2024:1-18. [PMID: 39091137 DOI: 10.1080/1040841x.2024.2384885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 05/25/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Frequent viral infections leading to infectious disease outbreaks have become a significant global health concern. Fully elucidating the molecular mechanisms of the immune response against viral infections is crucial for epidemic prevention and control. The innate immune response, the host's primary defense against viral infection, plays a pivotal role and has become a breakthrough in research mechanisms. A component of the innate immune system, damage-associated molecular patterns (DAMPs) are involved in inducing inflammatory responses to viral infections. Numerous DAMPs are released from virally infected cells, activating downstream signaling pathways via internal and external receptors on immune cells. This activation triggers immune responses and helps regulate viral host invasion. This review examines the immune regulatory mechanisms of various DAMPs, such as the S100 protein family, high mobility group box 1 (HMGB1), and heat shock proteins, in various viral infections to provide a theoretical basis for designing novel antiviral drugs.
Collapse
Affiliation(s)
- Huizhen Tian
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Qiong Liu
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Xiaomin Yu
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
- Medical Experimental Teaching Center, School of Basic Medical Sciences, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanli Cao
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- School of Basic Medical Sciences, Jiangxi medical College, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Yuan J, Guo L, Ma J, Zhang H, Xiao M, Li N, Gong H, Yan M. HMGB1 as an extracellular pro-inflammatory cytokine: Implications for drug-induced organic damage. Cell Biol Toxicol 2024; 40:55. [PMID: 39008169 PMCID: PMC11249443 DOI: 10.1007/s10565-024-09893-2] [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: 02/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024]
Abstract
Drug-induced organic damage encompasses various intricate mechanisms, wherein HMGB1, a non-histone chromosome-binding protein, assumes a significant role as a pivotal hub gene. The regulatory functions of HMGB1 within the nucleus and extracellular milieu are interlinked. HMGB1 exerts a crucial regulatory influence on key biological processes including cell survival, inflammatory regulation, and immune response. HMGB1 can be released extracellularly from the cell during these processes, where it functions as a pro-inflammation cytokine. HMGB1 interacts with multiple cell membrane receptors, primarily Toll-like receptors (TLRs) and receptor for advanced glycation end products (RAGE), to stimulate immune cells and trigger inflammatory response. The excessive or uncontrolled HMGB1 release leads to heightened inflammatory responses and cellular demise, instigating inflammatory damage or exacerbating inflammation and cellular demise in different diseases. Therefore, a thorough review on the significance of HMGB1 in drug-induced organic damage is highly important for the advancement of pharmaceuticals, ensuring their effectiveness and safety in treating inflammation as well as immune-related diseases. In this review, we initially outline the characteristics and functions of HMGB1, emphasizing their relevance in disease pathology. Then, we comprehensively summarize the prospect of HMGB1 as a promising therapeutic target for treating drug-induced toxicity. Lastly, we discuss major challenges and propose potential avenues for advancing the development of HMGB1-based therapeutics.
Collapse
Affiliation(s)
- JianYe Yuan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pathology, The Eight Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Lin Guo
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - JiaTing Ma
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - HeJian Zhang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - MingXuan Xiao
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Ning Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Hui Gong
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacy, Central South University, Changsha, China
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China
| | - Miao Yan
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha, China.
- Institute of Clinical Pharmacy, Central South University, Changsha, China.
- International Research Center for Precision Medicine, Transformative Technology and Software Services, Hunan, China.
| |
Collapse
|
9
|
Gao T, Huang Z. Effects of Isoflurane on the Cell Pyroptosis in the Lung Cancer Through the HMGB1/RAGE Pathway. Appl Biochem Biotechnol 2024; 196:3786-3799. [PMID: 37782455 DOI: 10.1007/s12010-023-04739-9] [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] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
There are many common malignant tumors in clinic. Among them, lung cancer is caused by the failure of suction system, which seriously threatens the life safety of patients. Recent studies have found that anesthetics have achieved certain efficacy in many cancers. Isoflurane, an inhaled anesthetic, is used in this study to explore whether it can prevent the lung cancer development. The A549 and H1299 were purchased. Cell viability was tested by CCK-8 experiment. Cell death and pyroptosis were analyzed by PI staining as well as flow cytometry. HMGB1 as well as RAGE protein levels were tested by Western blot. The same is true of pyroxin-related proteins. The HMGB1 as well as RAGE levels in the lung cancer tissues were determined by Western blot along with immunohistochemistry. Isoflurane treatment can reduce cell viability and promote cell pyroptosis. Additionally, the protein levels of cleaved caspase-1, IL-1β, GSDMD-N, NLRP3, HMGB1, and RAGE were dramatically up-regulated in the lung cancer after isoflurane treatment. Down-regulated proteins in lung cancer tissues include HMGB1 and RAGE proteins. After HMGB1 knockdown or FPS-ZM1 treatment, the role of isoflurane in the lung cancer was neutralized. This study demonstrated that isoflurane induced the cell pyroptosis in the lung cancer through activating the HMGB1/RAGE pathway.
Collapse
Affiliation(s)
- Tingting Gao
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Zeqing Huang
- Department of Anesthesia, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.
| |
Collapse
|
10
|
Chen C, Liu J, Lin X, Xiang A, Ye Q, Guo J, Rui T, Xu J, Hu S. Crosstalk between cancer-associated fibroblasts and regulated cell death in tumors: insights into apoptosis, autophagy, ferroptosis, and pyroptosis. Cell Death Discov 2024; 10:189. [PMID: 38649701 PMCID: PMC11035635 DOI: 10.1038/s41420-024-01958-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs), the main stromal component of the tumor microenvironment (TME), play multifaceted roles in cancer progression through paracrine signaling, exosome transfer, and cell interactions. Attractively, recent evidence indicates that CAFs can modulate various forms of regulated cell death (RCD) in adjacent tumor cells, thus involving cancer proliferation, therapy resistance, and immune exclusion. Here, we present a brief introduction to CAFs and basic knowledge of RCD, including apoptosis, autophagy, ferroptosis, and pyroptosis. In addition, we further summarize the different types of RCD in tumors that are mediated by CAFs, as well as the effects of these modes of RCD on CAFs. This review will deepen our understanding of the interactions between CAFs and RCD and might offer novel therapeutic avenues for future cancer treatments.
Collapse
Affiliation(s)
- Cong Chen
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Liu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Xia Lin
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Aizhai Xiang
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Qianwei Ye
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jufeng Guo
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Tao Rui
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Jian Xu
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Shufang Hu
- Department of Breast Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
11
|
Zhao P, Li Y, Xu X, Yang H, Li X, Fu S, Guo Z, Zhang J, Li H, Tian J. Neutrophil extracellular traps mediate cardiomyocyte ferroptosis via the Hippo-Yap pathway to exacerbate doxorubicin-induced cardiotoxicity. Cell Mol Life Sci 2024; 81:122. [PMID: 38456997 PMCID: PMC10923748 DOI: 10.1007/s00018-024-05169-4] [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: 10/12/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024]
Abstract
Doxorubicin-induced cardiotoxicity (DIC), which is a cardiovascular complication, has become the foremost determinant of decreased quality of life and mortality among survivors of malignant tumors, in addition to recurrence and metastasis. The limited ability to accurately predict the occurrence and severity of doxorubicin-induced injury has greatly hindered the prevention of DIC, but reducing the dose to mitigate side effects may compromise the effective treatment of primary malignancies. This has posed a longstanding clinical challenge for oncologists and cardiologists. Ferroptosis in cardiomyocytes has been shown to be a pivotal mechanism underlying cardiac dysfunction in DIC. Ferroptosis is influenced by multiple factors. The innate immune response, as exemplified by neutrophil extracellular traps (NETs), may play a significant role in the regulation of ferroptosis. Therefore, the objective of this study was to investigate the involvement of NETs in doxorubicin-induced cardiomyocyte ferroptosis and elucidate their regulatory role. This study confirmed the presence of NETs in DIC in vivo. Furthermore, we demonstrated that depleting neutrophils effectively reduced the occurrence of doxorubicin-induced ferroptosis and myocardial injury in DIC. Additionally, our findings showed the pivotal role of high mobility group box 1 (HMGB1) as a critical molecule implicated in DIC and emphasized its involvement in the modulation of ferroptosis subsequent to NETs inhibition. Mechanistically, we obtained preliminary evidence suggesting that doxorubicin-induced NETs could modulate yes-associated protein (YAP) activity by releasing HMGB1, which subsequently bound to toll like receptor 4 (TLR4) on the cardiomyocyte membrane, thereby influencing cardiomyocyte ferroptosis in vitro. Our findings suggest that doxorubicin-induced NETs modulate cardiomyocyte ferroptosis via the HMGB1/TLR4/YAP axis, thereby contributing to myocardial injury. This study offers a novel approach for preventing and alleviating DIC by targeting alterations in the immune microenvironment.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - You Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Xiangli Xu
- Department of Ultrasound, The Second Hospital of Harbin City, Harbin, 150001, China
| | - Haobo Yang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - Xintong Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- Ultrasound Molecular Imaging Joint Laboratory of Heilongjiang Province, Harbin Medical University, Harbin, 150001, China
| | - Shuai Fu
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Harbin, 150001, China
| | - Zihong Guo
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Jianing Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hairu Li
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
12
|
Fan A, Gao M, Tang X, Jiao M, Wang C, Wei Y, Gong Q, Zhong J. HMGB1/RAGE axis in tumor development: unraveling its significance. Front Oncol 2024; 14:1336191. [PMID: 38529373 PMCID: PMC10962444 DOI: 10.3389/fonc.2024.1336191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 02/15/2024] [Indexed: 03/27/2024] Open
Abstract
High mobility group protein 1 (HMGB1) plays a complex role in tumor biology. When released into the extracellular space, it binds to the receptor for advanced glycation end products (RAGE) located on the cell membrane, playing an important role in tumor development by regulating a number of biological processes and signal pathways. In this review, we outline the multifaceted functions of the HMGB1/RAGE axis, which encompasses tumor cell proliferation, apoptosis, autophagy, metastasis, and angiogenesis. This axis is instrumental in tumor progression, promoting tumor cell proliferation, autophagy, metastasis, and angiogenesis while inhibiting apoptosis, through pivotal signaling pathways, including MAPK, NF-κB, PI3K/AKT, ERK, and STAT3. Notably, small molecules, such as miRNA-218, ethyl pyruvate (EP), and glycyrrhizin exhibit the ability to inhibit the HMGB1/RAGE axis, restraining tumor development. Therefore, a deeper understanding of the mechanisms of the HMGB1/RAGE axis in tumors is of great importance, and the development of inhibitors targeting this axis warrants further exploration.
Collapse
Affiliation(s)
- Anqi Fan
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Mengxiang Gao
- College of Life Science, Yangtze University, Jingzhou, Hubei, China
| | - Xuhuan Tang
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengya Jiao
- Department of Immunology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonostic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chenchen Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
13
|
Liu T, Zhao H, Wang Y, Qu P, Wang Y, Wu X, Zhao T, Yang L, Mao H, Peng L, Zhan Y, Li P. Serum high mobility group box 1 as a potential biomarker for the progression of kidney disease in patients with type 2 diabetes. Front Immunol 2024; 15:1334109. [PMID: 38481996 PMCID: PMC10932975 DOI: 10.3389/fimmu.2024.1334109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Background As a damage-associated molecular pattern protein, high mobility group box 1 (HMGB1) is associated with kidney and systemic inflammation. The predictive and therapeutic value of HMGB1 as a biomarker has been confirmed in various diseases. However, its value in diabetic kidney disease (DKD) remains unclear. Therefore, this study aimed to investigate the correlation between serum and urine HMGB1 levels and DKD progression. Methods We recruited 196 patients with type 2 diabetes mellitus (T2DM), including 109 with DKD and 87 T2DM patients without DKD. Additionally, 60 healthy participants without T2DM were also recruited as controls. Serum and urine samples were collected for HMGB1 analysis. Simultaneously, tumor necrosis factor receptor superfamily member 1A (TNFR-1) in serum and kidney injury molecule (KIM-1) in urine samples were evaluated for comparison. Results Serum and urine HMGB1 levels were significantly higher in patients with DKD than in patients with T2DM and healthy controls. Additionally, serum HMGB1 levels significantly and positively correlated with serum TNFR-1 (R 2 = 0.567, p<0.001) and urine KIM-1 levels (R 2 = 0.440, p<0.001), and urine HMGB1 has a similar correlation. In the population with T2DM, the risk of DKD progression increased with an increase in serum HMGB1 levels. Multivariate logistic regression analysis showed that elevated serum HMGB1 level was an independent risk factor for renal function progression in patients with DKD, and regression analysis did not change in the model corrected for multiple variables. The restricted cubic spline depicted a nonlinear relationship between serum HMGB1 and renal function progression in patients with DKD (p-nonlinear=0.007, p<0.001), and this positive effect remained consistent across subgroups. Conclusion Serum HMGB1 was significantly correlated with DKD and disease severity. When the HMGB1 level was ≥27 ng/ml, the risk of renal progression increased sharply, indicating that serum HMGB1 can be used as a potential biomarker for the diagnosis of DKD progression.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hailing Zhao
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Ying Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Qu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yanmei Wang
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Xiai Wu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Tingting Zhao
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| |
Collapse
|
14
|
Zhu K, Fan R, Cao Y, Yang W, Zhang Z, Zhou Q, Ren J, Shi X, Gao Y, Guo X. Glycyrrhizin attenuates myocardial ischemia reperfusion injury by suppressing Inflammation, oxidative stress, and ferroptosis via the HMGB1-TLR4-GPX4 pathway. Exp Cell Res 2024; 435:113912. [PMID: 38176464 DOI: 10.1016/j.yexcr.2024.113912] [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: 09/17/2023] [Revised: 12/30/2023] [Accepted: 01/01/2024] [Indexed: 01/06/2024]
Abstract
Ferroptosis, a form of regulated cell death process, play an important role in myocardial ischemia‒reperfusion (I/R) injury. Glycyrrhizin (GL), a natural glycoconjugate triterpene, has the property to improve growth rate, immune regulation, antioxidant, anti-inflammatory. However, whether GL can attenuate myocardial I/R injury by modulating ferroptosis or other mechanisms are still unclear. In this study, SD rats underwent in vivo myocardial ischemia/reperfusion (I/R) surgery, while H9C2 cells were subjected to the hypoxia/reoxygenation (H/R) model for in vitro experiments. In addition, TAK-242, a TLR4-specific antagonist, and GL were also used to evaluate the effect and mechanisms of GL on the cardiac function and expression of ferroptosis-related gene and protein in vivo and vitro. The results show that GL decreased not only the expression of the inflammation-related factors (HMGB1, TNF-α, IL-6, IL-18 and IL-1β), but also reduced the number of TUNEL-positive cardiomyocytes, and mitigated pathological alterations in I/R injury. In addition, GL decreased the levels of MDA, promoted antioxidant capacity such as GSH, CAT, Cu/Zn-SOD, Mn-SOD, and SOD in vivo and vitro. More importantly, GL and TAK-242 regulate ferroptosis-related protein and gene expression in I/R and H/R model. Surprisingly, GL may ameliorate cardiomyocyte ferroptosis and ultimately improves cardiac function induced by H/R via the HMGB1-TLR4-GPX4 axis. Therefore, we have highlighted a novel mechanism by which GL regulates inflammation, oxidative stress, and ferroptosis via the HMGB1-TLR4-GPX4 pathway to prevent myocardial I/R injury. GL appears to be a potentially applicable drug for the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Kaiyi Zhu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China.
| | - Rong Fan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuchen Cao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yang
- Department of Neurosurgery, Beijing Children's Hospital, Capital Medical University, Beijing, 100045, China
| | - Zhe Zhang
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, 030032, China; Department of Pulmonary and Critical Care Medicine, Aerospace Center Hospital, Beijing, 100049, China
| | - Qiang Zhou
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Ren
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiushan Shi
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cellular Physiology, Shanxi Province, Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, China.
| | - Xiang Guo
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
15
|
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
|
16
|
Zhang YJ, Huang C, Zu XG, Liu JM, Li YJ. Use of Machine Learning for the Identification and Validation of Immunogenic Cell Death Biomarkers and Immunophenotypes in Coronary Artery Disease. J Inflamm Res 2024; 17:223-249. [PMID: 38229693 PMCID: PMC10790656 DOI: 10.2147/jir.s439315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/28/2023] [Indexed: 01/18/2024] Open
Abstract
Objective Immunogenic cell death (ICD) is part of the immune system's response to coronary artery disease (CAD). In this study, we bioinformatically evaluated the diagnostic and therapeutic utility of immunogenic cell death-related genes (IRGs) and their relationship with immune infiltration features in CAD. Methods We acquired the CAD-related datasets GSE12288, GSE71226, and GSE120521 from the Gene Expression Omnibus (GEO) database and the IRGs from the GeneCards database. After identifying the immune cell death-related differentially expressed genes (IRDEGs), we developed a risk model and detected immune subtypes in CAD. IRDEGs were identified using least absolute shrinkage and selection operator (LASSO) analysis. Using a nomogram, we confirmed that both the LASSO model and ICD signature genes had good diagnostic performance. Results There was a high degree of coincidence and immune representativeness between two CAD groups based on characteristic genes and hub genes. Hub genes were associated with the interaction of neuroactive ligands with receptors and cell adhesion receptors. The two groups differed in terms of adipogenesis, allograft rejection, and apoptosis, as well as the ICD signature and hub gene expression levels. The two CAD-ICD subtypes differed in terms of immune infiltration. Conclusion Quantitative real-time PCR (qRT-PCR) correlated CAD with the expression of OAS3, ITGAV, and PIBF1. The ICD signature genes are candidate biomarkers and reference standards for immune grouping in CAD and can be beneficial in precise immune-targeted therapy.
Collapse
Affiliation(s)
- Yan-jiao Zhang
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Chao Huang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Xiu-guang Zu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Jin-ming Liu
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| | - Yong-jun Li
- Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, People’s Republic of China
| |
Collapse
|
17
|
Yang X, Dong X, Li J, Zheng A, Shi W, Shen C, Liu J. Nanocurcumin attenuates pyroptosis and inflammation through inhibiting NF-κB/GSDMD signal in high altitude-associated acute liver injury. J Biochem Mol Toxicol 2024; 38:e23606. [PMID: 38050447 DOI: 10.1002/jbt.23606] [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: 05/07/2022] [Revised: 07/12/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023]
Abstract
Exposure to a hypobaric hypoxic environment at high altitudes can lead to liver injury, and mounting evidence indicates that pyroptosis and inflammation play important roles in liver injury. Curcumin (Cur) can inhibit pyroptosis and inflammation. Therefore, our purpose here was to clarify the mechanism underlying the protective effect of nanocurcumin (Ncur) and Cur in a rat model of high altitude-associated acute liver injury. Eighty healthy rats were selected and exposed to different altitudes (6000 or 7000 m) for 0, 24, 48, or 72 h. Fifty normal healthy rats were divided into normal control, high-altitude control, salidroside (40 mg/kg [Sal-40]), Cur (200 mg/kg [Cur-200]), and Ncur (25 mg/kg [Ncur-25]) groups and exposed to a high-altitude hypobaric hypoxic environment (48 h, 7000 m). Serum-liver enzyme activities (alanine transaminase, aspartate transaminase, and lactate dehydrogenase were detected and histopathology of liver injury was evaluated by hematoxylin and eosin staining, and inflammatory factors were detected in liver tissues by enzyme-linked immunosorbent assays. Pyroptosis-associated proteins (gasdermin D, gasdermin D N-terminal [GSDMD-N], pro-Caspase-1, and cleaved-Caspase-1 [cleaved-Casp1]) and inflammation-associated proteins (nuclear factor-κB [NF-κB], phospho-NF-κB [P-NF-κB], and high-mobility group protein B1 [HMGB1]) levels were analyzed by immunoblotting. Ncur and Cur inhibited increased serum-liver enzyme activities, alleviated liver injury in rats caused by high-altitude hypobaric hypoxic exposure, and downregulated inflammatory factors, including tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and IL-18, in rat liver tissues. The level of P-NF-κB, GSDMD-N, cleaved-Casp1, and HMGB1 in rat liver tissues increased significantly after high-altitude exposure. Ncur and Cur downregulated P-NF-κB, GSDMD-N, cleaved-Casp-1, and HMGB1. Ncur and Cur may inhibit inflammatory responses and pyroptosis in a rat model of high altitude-associated acute liver injury.
Collapse
Affiliation(s)
- Xinyue Yang
- Key Laboratory of Special Environmental Medicine of Xinjiang, General Hospital of Xinjang Military Command, Urumqi, China
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Xiang Dong
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Jiajia Li
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Aiping Zheng
- Institute of Pharmacology and Toxicology, Academy of Military Medicine, Beijing, China
| | - Wenhui Shi
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Caifu Shen
- Graduate School, Xinjiang Medical University, Urumqi, China
| | - Jiangwei Liu
- Graduate School, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
18
|
Li S, Tao G. Perish in the Attempt: Regulated Cell Death in Regenerative and Nonregenerative Tissue. Antioxid Redox Signal 2023; 39:1053-1069. [PMID: 37218435 PMCID: PMC10715443 DOI: 10.1089/ars.2022.0166] [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: 10/05/2022] [Revised: 05/12/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Significance: A cell plays its roles throughout its life span, even during its demise. Regulated cell death (RCD) is one of the key topics in modern biomedical studies. It is considered the main approach for removing stressed and/or damaged cells. Research during the past two decades revealed more roles of RCD, such as coordinating tissue development and driving compensatory proliferation during tissue repair. Recent Advances: Compensatory proliferation, initially identified in primitive organisms during the regeneration of lost tissue, is an evolutionarily conserved process that also functions in mammals. Among various types of RCD, apoptosis is considered the top candidate to induce compensatory proliferation in damaged tissue. Critical Issues: The roles of apoptosis in the recovery of nonregenerative tissue are still vague. The roles of other types of RCD, such as necroptosis and ferroptosis, have not been well characterized in the context of tissue regeneration. Future Directions: In this review article, we attempt to summarize the recent insights on the role of RCD in tissue repair. We focus on apoptosis, with expansion to ferroptosis and necroptosis, in primitive organisms with significant regenerative capacity as well as common mammalian research models. After gathering hints from regenerative tissue, in the second half of the review, we take a notoriously nonregenerative tissue, the myocardium, as an example to discuss the role of RCD in terminally differentiated quiescent cells. Antioxid. Redox Signal. 39, 1053-1069.
Collapse
Affiliation(s)
- Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
19
|
Yan W, Tang HY, Yang YQ, He KL. Serum uric acid and outcome in hospitalized elderly patients with chronic heart failure through the whole spectrum of ejection fraction phenotypes. BMC Cardiovasc Disord 2023; 23:589. [PMID: 38036986 PMCID: PMC10688460 DOI: 10.1186/s12872-023-03544-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/04/2023] [Indexed: 12/02/2023] Open
Abstract
INTRODUCTION Elevated serum uric acid (SUA) levels have been associated with poor outcome in patients with heart failure (HF). Uric acid is associated with inflammation and microvascular dysfunction, which may differentially affect left ventricular ejection fraction (EF) phenotypes. We aimed to identify the role of SUA across EF phenotypes in hospitalized elderly patients with chronic HF. METHODS We analyzed 1355 elderly patients who were diagnosed with chronic HF. All patients had SUA levels measured within the first 24 h following admission. Patients with left ventricle EF were categorized as having HF with reduced EF (HFrEF, EF < 40%), HF with mid-range EF (HFmrEF, 40%≦LVEF ≦ 49%) or HF with preserved EF (HFpEF, LVEF ≥ 50%). Endpoints were cardiovascular death, HF rehospitalization, and their composite. The median follow-up period was 18 months. RESULTS Compared with the lowest SUA quartile, the highest SUA quartile was significantly associated with the endpoints (adjusted HR: 2.404, 95% CI: 1.178-4.906, P = 0.016; HR: 1.418, 95% CI: 1.021-1.971, P = 0.037; HR: 1.439, 95% CI: 1.049-1.972, P = 0.024, respectively). After model adjustment, a significant association of SUA with cardiovascular death and the composite endpoint persisted among HFrEF and HFmrEF patients in the highest SUA quartile (P < 0.05 for all). CONCLUSIONS In hospitalized elderly patients with chronic HF, SUA is an independent predictor of adverse outcomes, which can be seen in HFrEF and HFmrEF patients.
Collapse
Affiliation(s)
- Wei Yan
- Department of Geriatric Medicine, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China
| | - Hai-Ying Tang
- Department of Geriatric Medicine, The First Affiliated Hospital of Soochow University, 188 Shizijie Road, Suzhou, 215006, Jiangsu, China.
| | - Yong-Qiang Yang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, San Xiang Road No. 1055, Suzhou, 215004, Jiangsu, China.
| | - Kun-Lun He
- Analysis Center of Big Data, Medical Innovation Research Division, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
20
|
He J, Zhang Q, Xia X, Yang L. Lagopsis supina ameliorates myocardial ischemia injury by regulating angiogenesis, thrombosis, inflammation, and energy metabolism through VEGF, ROS and HMGB1 signaling pathways in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 120:155050. [PMID: 37708818 DOI: 10.1016/j.phymed.2023.155050] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/14/2023] [Accepted: 08/24/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Lagopsis supina (Steph. ex. Willd.) Ikonn.-Gal. is an important traditional Chinese medicine used to treat various ailments. However, its impact on myocardial ischemia (MI) injury remains unknown. PURPOSE This research aimed to reveal the therapeutic effect, potential mechanism, and metabolomics of L. supina against MI injury in rats. METHODS The therapeutic effects of the ethanolic extract of L. supina (LS) and its four fractions (LSA∼D) on a left anterior descending (LAD) artery occlusion-induced MI model rat were explored. The pharmacodynamics including myocardial infraction area, myocardial tissue pathology and apoptosis, and serum biochemical parameters (CK, CK-MB, CTn-T, SOD, ET-1, NO, eNOS, VEGF, TXB2, 6-keto-PGF1α, TNF-α, IL-6, and CRP) were evaluated. The 24 related protein expressions were detected using western blotting assay. Simultaneously, the qualitative and quantitative analyses of microporous adsorption resin with 30% (LSC) and 60% (LSD) aqueous ethanol fractions were performed using UHPLC-MS and HPLC. Moreover, the serum metabolomics analysis of rats was profiled using UHPLC-MS. RESULTS LS exerted remarkable alleviating effect on MI in rats. Importantly, LSC and LSD, two effective fractions of LS, significantly reduced myocardial infraction area, alleviated myocardial tissue pathology and apoptosis, regulated serum biochemical parameters. Furthermore, LSC and LSD markedly up-regulated the levels of VEGF-A, VEGFR-2, PKC, Bcl-2, Nrf2, HO-1, and thrombin, as well as prominently down-regulated the protein expression of Notch 1, p-PI3K, p-PI3K/PI3K, p-Akt, p-Akt/Akt, Bax, cleaved-caspase-3, cleaved-caspase-3/caspase-3, vWF, p-Erk, p-Erk/Erk, HMGB1, p-p38, p-p38/p38, p-p65, and p-p65/p65. A total of 26 candidate biomarkers were significantly regulated by LSC and LSD and they are mainly involved in amino acid metabolism, glycerophospholipid metabolism, and sphingolipid metabolism. Finally, phenylethanols and flavonoids may be major bio-constituents of LSC and LSD against MI. CONCLUSIONS This work, for the first time, demonstrated that L. supina had a significant therapeutic effect on MI in rats. Additionally, LSC and LSD, two bio-fractions from L. supina, exerted their potential to ameliorate MI injury by promoting angiogenesis, inhibiting thrombosis, blocking inflammation, and facilitating energy metabolism through promotion of VEGF pathway, as well as suppression of ROS and HMGB1 pathways in rats. These findings suggest that LSC and LSD hold promise as potential therapeutic agents for MI injury in clinical application.
Collapse
Affiliation(s)
- Junwei He
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qingcui Zhang
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China; College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Xiaoyi Xia
- Research Center of Natural Resources of Chinese Medicinal Materials and Ethnic Medicine, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Li Yang
- College of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang 330004, China.
| |
Collapse
|
21
|
Chen X, Liu Q, Wu E, Ma Z, Tuo B, Terai S, Li T, Liu X. The role of HMGB1 in digestive cancer. Biomed Pharmacother 2023; 167:115575. [PMID: 37757495 DOI: 10.1016/j.biopha.2023.115575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023] Open
Abstract
High mobility group box protein B1 (HMGB1) belongs to the HMG family, is widely expressed in the nucleus of digestive mucosal epithelial cells, mesenchymal cells and immune cells, and binds to DNA to participate in genomic structural stability, mismatch repair and transcriptional regulation to maintain normal cellular activities. In the context of digestive inflammation and tumors, HMGB1 readily migrates into the extracellular matrix and binds to immune cell receptors to affect their function and differentiation, further promoting digestive tract tissue injury and tumor development. Notably, HMGB1 can also promote the antitumor immune response. Therefore, these seemingly opposing effects in tumors make targeted HMGB1 therapies important in digestive cancer. This review focuses on the role of HMGB1 in tumors and its effects on key pathways of digestive cancer and aims to provide new possibilities for targeted tumor therapy.
Collapse
Affiliation(s)
- Xiangqi Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Qian Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Enqing Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Shuji Terai
- Division of Gastroenterology & Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Japan
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
22
|
Wang K, Li Y. Signaling pathways and targeted therapeutic strategies for polycystic ovary syndrome. Front Endocrinol (Lausanne) 2023; 14:1191759. [PMID: 37929034 PMCID: PMC10622806 DOI: 10.3389/fendo.2023.1191759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 09/18/2023] [Indexed: 11/07/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is the most common endocrine disorder among women of reproductive age. Although promising strides have been made in the field of PCOS over the past decades, the distinct etiologies of this syndrome are not fully elucidated. Prenatal factors, genetic variation, epigenetic mechanisms, unhealthy lifestyles, and environmental toxins all contribute to the development of this intricate and highly heterogeneous metabolic, endocrine, reproductive, and psychological disorder. Moreover, interactions between androgen excess, insulin resistance, disruption to the hypothalamic-pituitary-ovary (HPO) axis, and obesity only make for a more complex picture. In this review, we investigate and summarize the related molecular mechanisms underlying PCOS pathogenesis from the perspective of the level of signaling pathways, including PI3K/Akt, TGF-β/Smads, Wnt/β-catenin, and Hippo/YAP. Additionally, this review provides an overview of prospective therapies, such as exosome therapy, gene therapy, and drugs based on traditional Chinese medicine (TCM) and natural compounds. By targeting these aberrant pathways, these interventions primarily alleviate inflammation, insulin resistance, androgen excess, and ovarian fibrosis, which are typical symptoms of PCOS. Overall, we hope that this paper will pave the way for better understanding and management of PCOS in the future.
Collapse
Affiliation(s)
- Kexin Wang
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanhua Li
- Department of General Practice, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
23
|
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: 5] [Impact Index Per Article: 2.5] [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
|
24
|
Millán Solano MV, Salinas Lara C, Sánchez-Garibay C, Soto-Rojas LO, Escobedo-Ávila I, Tena-Suck ML, Ortíz-Butrón R, Choreño-Parra JA, Romero-López JP, Meléndez Camargo ME. Effect of Systemic Inflammation in the CNS: A Silent History of Neuronal Damage. Int J Mol Sci 2023; 24:11902. [PMID: 37569277 PMCID: PMC10419139 DOI: 10.3390/ijms241511902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 08/13/2023] Open
Abstract
Central nervous system (CNS) infections including meningitis and encephalitis, resulting from the blood-borne spread of specific microorganisms, provoke nervous tissue damage due to the inflammatory process. Moreover, different pathologies such as sepsis can generate systemic inflammation. Bacterial lipopolysaccharide (LPS) induces the release of inflammatory mediators and damage molecules, which are then released into the bloodstream and can interact with structures such as the CNS, thus modifying the blood-brain barrier's (BBB´s) and blood-cerebrospinal fluid barrier´s (BCSFB´s) function and inducing aseptic neuroinflammation. During neuroinflammation, the participation of glial cells (astrocytes, microglia, and oligodendrocytes) plays an important role. They release cytokines, chemokines, reactive oxygen species, nitrogen species, peptides, and even excitatory amino acids that lead to neuronal damage. The neurons undergo morphological and functional changes that could initiate functional alterations to neurodegenerative processes. The present work aims to explain these processes and the pathophysiological interactions involved in CNS damage in the absence of microbes or inflammatory cells.
Collapse
Affiliation(s)
- Mara Verónica Millán Solano
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cos’ıo Villegas, Mexico City 14080, Mexico;
| | - Citlaltepetl Salinas Lara
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Carlos Sánchez-Garibay
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Luis O. Soto-Rojas
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Itzel Escobedo-Ávila
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Departamento de Neurodesarrollo y Fisiología, Instituto de Fisiología Celular, Universidad Nacional Autonoma de Mexico, Mexico City 04510, Mexico
| | - Martha Lilia Tena-Suck
- Departamento de Neuropatología, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suarez, Mexico City 14269, Mexico;
| | - Rocío Ortíz-Butrón
- Laboratorio de Neurobiología, Departamento de Fisiología de ENCB, Instituto Politécnico Nacional, Mexico City 07738, Mexico;
| | - José Alberto Choreño-Parra
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cos’ıo Villegas, Mexico City 14080, Mexico;
| | - José Pablo Romero-López
- Red MEDICI, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de Mexico, Tlalnepantla 54090, Mexico; (M.V.M.S.); (C.S.-G.); (L.O.S.-R.); (I.E.-Á.); (J.P.R.-L.)
- Laboratorio de Patogénesis Molecular, Laboratorio 4, Edificio A4, Carrera Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - María Estela Meléndez Camargo
- Laboratorio de Farmacología, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu Esq. Manuel Luis Stampa S/N, U.P. Adolfo López Mateos, Mexico City 07738, Mexico;
| |
Collapse
|
25
|
Mo J, Hu J, Cheng X. The role of high mobility group box 1 in neuroinflammatory related diseases. Biomed Pharmacother 2023; 161:114541. [PMID: 36963363 DOI: 10.1016/j.biopha.2023.114541] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/12/2023] [Accepted: 03/13/2023] [Indexed: 03/26/2023] Open
Abstract
High mobility group box 1 (HMGB1) is a ubiquitous and highly conserved non-histone DNA-binding protein with different biological functions according to its subcellular localization. It is widely believed that HMGB1, which is released into the extracellular space, plays a key role in the inflammatory response. In recent years, numerous studies have shown that the development of various neurological diseases such as epilepsy, Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), cerebrovascular disease and traumatic brain injury (TBI) are inextricably linked to inflammation. We will review the mechanisms of HMGB1 and its receptors in nervous system inflammation to provide a basis for further development of new HMGB1-based therapies.
Collapse
Affiliation(s)
- Jialu Mo
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China
| | - Jiao Hu
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China
| | - Xianglin Cheng
- The First Affiliated Hospital of Yangtze University, Jingzhou 434000, Hubei, China.
| |
Collapse
|
26
|
Gauthier AG, Lin M, Zefi S, Kulkarni A, Thakur GA, Ashby CR, Mantell LL. GAT107-mediated α7 nicotinic acetylcholine receptor signaling attenuates inflammatory lung injury and mortality in a mouse model of ventilator-associated pneumonia by alleviating macrophage mitochondrial oxidative stress via reducing MnSOD-S-glutathionylation. Redox Biol 2023; 60:102614. [PMID: 36717349 PMCID: PMC9950665 DOI: 10.1016/j.redox.2023.102614] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Supraphysiological concentrations of oxygen (hyperoxia) can compromise host defense and increase susceptibility to bacterial and viral infections, causing ventilator-associated pneumonia (VAP). Compromised host defense and inflammatory lung injury are mediated, in part, by high extracellular concentrations of HMGB1, which can be decreased by GTS-21, a partial agonist of α7 nicotinic acetylcholine receptor (α7nAChR). Here, we report that a novel α7nAChR agonistic positive allosteric modulator (ago-PAM), GAT107, at 3.3 mg/kg, i.p., significantly decreased animal mortality and markers of inflammatory injury in mice exposed to hyperoxia and subsequently infected with Pseudomonas aeruginosa. The incubation of macrophages with 3.3 μM of GAT107 significantly decreased hyperoxia-induced extracellular HMGB1 accumulation and HMGB1-induced macrophage phagocytic dysfunction. Hyperoxia-compromised macrophage function was correlated with impaired mitochondrial membrane integrity, increased superoxide levels, and decreased manganese superoxide dismutase (MnSOD) activity. This compromised MnSOD activity is due to a significant increase in its level of glutathionylation. The incubation of hyperoxic macrophages with 3.3 μM of GAT107 significantly decreases the levels of glutathionylated MnSOD, and restores MnSOD activity and mitochondrial membrane integrity. Thus, GAT107 restored hyperoxia-compromised phagocytic functions by decreasing HMGB1 release, most likely via a mitochondrial-directed pathway. Overall, our results suggest that GAT107 may be a potential treatment to decrease acute inflammatory lung injury by increasing host defense in patients with VAP.
Collapse
Affiliation(s)
- Alex G. Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Sidorela Zefi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | | | | | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA,Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, USA,Corresponding author. Department of Pharmaceutical Sciences, St. John's University College of Pharmacy and Health Sciences, 128 St. Albert Hall, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
27
|
Zhang M, Lin Y, Chen R, Yu H, Li Y, Chen M, Dou C, Yin P, Zhang L, Tang P. Ghost messages: cell death signals spread. Cell Commun Signal 2023; 21:6. [PMID: 36624476 PMCID: PMC9830882 DOI: 10.1186/s12964-022-01004-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 11/24/2022] [Indexed: 01/11/2023] Open
Abstract
Cell death is a mystery in various forms. Whichever type of cell death, this is always accompanied by active or passive molecules release. The recent years marked the renaissance of the study of these molecules showing they can signal to and communicate with recipient cells and regulate physio- or pathological events. This review summarizes the defined forms of messages cells could spread while dying, the effects of these signals on the target tissue/cells, and how these types of communications regulate physio- or pathological processes. By doing so, this review hopes to identify major unresolved questions in the field, formulate new hypothesis worthy of further investigation, and when possible, provide references for the search of novel diagnostic/therapeutics agents. Video abstract.
Collapse
Affiliation(s)
- Mingming Zhang
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Yuan Lin
- grid.412463.60000 0004 1762 6325Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001 Heilongjiang People’s Republic of China
| | - Ruijing Chen
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Haikuan Yu
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Yi Li
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Ming Chen
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Ce Dou
- grid.410570.70000 0004 1760 6682Department of Orthopedics, Southwest Hospital, Army Medical University, Chongqing, 400038 People’s Republic of China
| | - Pengbin Yin
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Licheng Zhang
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| | - Peifu Tang
- grid.414252.40000 0004 1761 8894Department of Orthopedics, Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China ,National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Beijing, 100853 People’s Republic of China
| |
Collapse
|
28
|
Tian Y, Chen R, Su Z. HMGB1 is a Potential and Challenging Therapeutic Target for Parkinson's Disease. Cell Mol Neurobiol 2023; 43:47-58. [PMID: 34797463 PMCID: PMC11415213 DOI: 10.1007/s10571-021-01170-8] [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: 07/14/2021] [Accepted: 11/14/2021] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is one of the most common degenerative diseases of the human nervous system and has a wide range of serious impacts on human health and quality of life. Recently, research targeting high mobility group box 1 (HMGB1) in PD has emerged, and a variety of laboratory methods for inhibiting HMGB1 have achieved good results to a certain extent. However, given that HMGB1 undergoes a variety of intracellular modifications and three different forms of extracellular redox, the possible roles of these forms in PD are likely to be different. General inhibition of all forms of HMGB1 is obviously not ideal and has become one of the biggest obstacles in the clinical application of targeting HMGB1. In this review, pure mechanistic research of HMGB1 and in vivo research targeting HMGB1 were combined, the effects of HMGB1 on neurons and immune cell responses in PD are discussed in detail, and the problems that need to be focused on in the future are addressed.
Collapse
Affiliation(s)
- Yu Tian
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Rong Chen
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
29
|
Zhang J, Hu Y, Wang H, Hou J, Xiao W, Wen X, Wang T, Long P, Jiang H, Wang Z, Liu H, Chen X. Advances in research on the protective mechanisms of traditional Chinese medicine (TCM) in myocardial ischaemia-reperfusion injury. PHARMACEUTICAL BIOLOGY 2022; 60:931-948. [PMID: 35587352 PMCID: PMC9132412 DOI: 10.1080/13880209.2022.2063342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 01/31/2022] [Accepted: 03/31/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Developing effective drugs to treat myocardial ischaemia-reperfusion (MI/R) injury is imperative. Traditional Chinese medicines (TCMs) have had considerable success in the treatment of cardiovascular diseases. Elucidating the mechanisms by which TCMs improve MI/R injury can supplement the literature on MI/R prevention and treatment. OBJECTIVE To summarise TCMs and their main protective mechanisms against MI/R injury reported over the past 40 years. METHODS Relevant literature published between 1980 and 2020 in Chinese and English was retrieved from the Web of Science, PubMed, SpringerLink, PubMed Central, Scopus, and Chinese National Knowledge Infrastructure (CNKI) databases. Search terms included 'medicinal plants', 'myocardial ischaemia reperfusion injury', 'Chinese medicine prescriptions', 'mechanisms', 'prevention', 'treatment' and 'protection'. For inclusion in the analysis, medicinal plants had to be searchable in the China Medical Information Platform and Plant Database. RESULTS We found 71 medicinal species (from 40 families) that have been used to prevent MI/R injury, of which Compositae species (8 species) and Leguminosae species (7 species) made up the majority. Most of the effects associated with these plants are described as antioxidant and anti-inflammatory. Furthermore, we summarised 18 kinds of Chinese compound prescriptions, including the compound Danshen tablet and Baoxin pill, which mainly reduce oxidative stress and regulate mitochondrial energy metabolism. DISCUSSION AND CONCLUSIONS We summarised TCMs that protect against MI/R injury and their pharmacological mechanisms. This in-depth explanation of the roles of TCMs in MI/R injury protection provides a theoretical basis for the research and development of TCM-based treatment drugs.
Collapse
Affiliation(s)
- Jiexin Zhang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Yonghe Hu
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Han Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Wenjing Xiao
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Xudong Wen
- Department of Gastroenterology, The First People’s Hospital of Chengdu, Chengdu, Sichuan, China
| | - Tingting Wang
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Pan Long
- Department of Central Laboratory, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Hezhong Jiang
- Faculty of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Zhanhao Wang
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Huawei Liu
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| | - Xin Chen
- Department of Laboratory Medicine, The Third People’s Hospital of Chengdu/Affiliated Hospital of Southwest, Jiaotong University, Chengdu, Sichuan, China
| |
Collapse
|
30
|
Frustaci A, Russo MA, Chimenti C. Individualized immunosuppression in virus-negative inflammatory cardiomyopathy. Eur Heart J 2022; 43:4760. [PMID: 36263787 DOI: 10.1093/eurheartj/ehac559] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Andrea Frustaci
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, La Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy.,Cellular and Molecular Cardiology Lab, IRCCS L. Spallanzani, 00149 Rome, Italy
| | - Matteo A Russo
- MEBIC Consortium, San Raffaele Open University and IRCCS San Raffaele, 00166 Rome, Italy
| | - Cristina Chimenti
- Department of Clinical, Internal, Anesthesiologist and Cardiovascular Sciences, La Sapienza University, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
31
|
Bai J, Wu B, Zhao S, Wang G, Su S, Lu B, Hu Y, Geng Y, Guo Z, Wan J, OuYang W, Hu C, Liu J. The Effect of PD-1 Inhibitor Combined with Irradiation on HMGB1-Associated Inflammatory Cytokines and Myocardial Injury. J Inflamm Res 2022; 15:6357-6371. [PMID: 36424918 PMCID: PMC9680686 DOI: 10.2147/jir.s384279] [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: 09/01/2022] [Accepted: 11/08/2022] [Indexed: 10/17/2023] Open
Abstract
PURPOSE To explore the effect of PD-1 inhibitors combined with irradiation on myocardial injury and the changes of HMGB1-associated inflammatory markers. METHODS Four groups of five mice were used, each groupformed by randomly dividing 20 mice (group A control; group B PD-1 inhibitors; group C Irradiation; group D PD-1 inhibitors+irradiation; n = 5 for each). The mice were treated with either PD-1 inhibitors or a 15 Gy dose of single heart irradiation, or both. Hematoxylin-eosin staining assessed the morphology and pathology of heart tissue; Masson staining assessed heart fibrosis; Tunel staining evaluated heart apoptosis; flow cytometry detected CD3+, CD4+, and CD8+ T lymphocytes in heart tissues; enzyme linked immunosorbent assay evaluated IL-1β, IL-6, and TNF-ɑ of heart tissue; Western blot and quantitative real-time PCR (qPCR) detected the expression of protein and mRNA of HMGB1, TLR-4, and NF-κB p65 respectively. RESULTS The degree of heart injury, collagen volume fraction (CVF) and apoptotic index (AI) in groups B, C, and D were higher than group A, but the differences between the CVF and AI of group A and group B were not statistical significance (P>0.05). Similarly, the absolute counts and relative percentage of CD3+ and CD8+ T lymphocytes and the concentrations of IL-1β, IL-6, and TNF-α in heart tissue with group D were significantly higher than the other groups (P<0.05). In addition, compared with group A, the expression of protein and mRNA of HMGB1 and NF-κB p65 in other groups were higher, and the differences between each group were statistically significant while TLR4 was not. In addition, interaction by PD-1 inhibitors and irradiation was found in inflammatory indicators, especially in the expression of the HMGB1 and CD8+ T lymphocytes. CONCLUSION PD-1 inhibitors can increase the expression of HMGB1-associated inflammatory cytokines and aggravate radiation-induced myocardial injury.
Collapse
Affiliation(s)
- Jie Bai
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Bibo Wu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Shasha Zhao
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Gang Wang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Shengfa Su
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Bing Lu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Yinxiang Hu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Yichao Geng
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Zhengneng Guo
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Jun Wan
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Weiwei OuYang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Cheng Hu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| | - Jie Liu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, People’s Republic of China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, People’s Republic of China
| |
Collapse
|
32
|
Luo L, Wang S, Chen B, Zhong M, Du R, Wei C, Huang F, Kou X, Xing Y, Tong G. Inhibition of inflammatory liver injury by the HMGB1-A box through HMGB1/TLR-4/NF-κB signaling in an acute liver failure mouse model. Front Pharmacol 2022; 13:990087. [PMID: 36313316 PMCID: PMC9614247 DOI: 10.3389/fphar.2022.990087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
We aimed to investigate the preventive effect of high mobility group box 1 (HMGB1)-A box and the mechanism by which it alleviates inflammatory injury in acute liver failure (ALF) by inhibiting the extracellular release of HMGB1. BALB/c mice were intraperitoneally (i.p.) administered LPS/D-GalN to establish an ALF mouse model. HMGB1-A box was administered (i.p.) 1 h before establishing the ALF mouse model. The levels of extracellularly released HMGB1, TLR-4/NF-κB signaling molecules, the proinflammatory cytokines TNF-α, IL-1β, and IL-6 and COX-2 were measured in the liver tissue and/or serum by Immunohistochemistry, Western blotting and Enzyme-linked immunosorbent assay (ELISA). The levels of extracellularly released HMGB1, TLR-4/NF-κB signaling molecules and proinflammatory cytokines were measured in Huh7 cells as well as LPS- and/or HMGB1-A box treatment by confocal microscopy, Western blotting and ELISA. In the ALF mouse model, the levels of HMGB1 were significantly increased both in the liver and serum, TLR-4/NF-κB signaling molecules and proinflammatory cytokines also was upregulated. Notably, HMGB1-A box could reverse these changes. HMGB1-A box could also cause these changes in LPS-induced Huh7 cells. HMGB1-A box played a protective role by inhibiting inflammatory liver injury via the regulation of HMGB1/TLR-4/NF-κB signaling in the LPS/D-GaIN-induced ALF mouse model, which may be related to inhibiting the extracellular release of HMGB1.
Collapse
Affiliation(s)
- Lidan Luo
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Shuai Wang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Bohao Chen
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, China
| | - Mei Zhong
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, China
| | - Ruili Du
- Department of Hepatology, Shenzhen Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen, China
| | - ChunShan Wei
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Furong Huang
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| | - Xinhui Kou
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Yufeng Xing
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
| | - Guangdong Tong
- Department of Hepatology, The Fourth Clinical Medical College of Guangzhou University of Traditional Chinese Medicine, Shenzhen, China
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, China
| |
Collapse
|
33
|
Soltysova M, Tomova A, Ostatnikova D. Gut Microbiota Profiles in Children and Adolescents with Psychiatric Disorders. Microorganisms 2022; 10:2009. [PMID: 36296284 PMCID: PMC9608804 DOI: 10.3390/microorganisms10102009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/28/2022] [Accepted: 09/30/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of our work is to summarize the current state of knowledge on gut microbiota differences in children and adolescents with psychiatric disorders. To find the relevant articles, the PubMed, Web of Science, and Google Scholar databases were searched. Articles in English presenting original data and comparing the composition of gut microbiota in child psychiatric patients with gut microbiota in healthy children and adolescents were selected. Finally, we identified 55 articles eligible for our purpose. The majority of patients with autism spectrum disorders (ASD) were investigated. A smaller number of studies evaluating the gut microbiota in children and adolescents with attention-deficit/hyperactivity disorder (ADHD), Rett syndrome, anorexia nervosa, depressive disorder (DD), and tic disorders were found. The main findings of this research are discussed in our review, focusing on the age-related gut microbiota specificity for psychiatric disorders and the differences between individual diagnosis. To conclude, the gut microbiota in children and adolescents with psychiatric disorders is evidently different from that in controls. The most pronounced differences are seen in children with ASD, less in ADHD. Moreover, the changes are not identical to those in adult psychiatric patients, as Ruminococcus, Turicibacter, and Bilophila were increased in adults, and decreased in children with ASD, and Parabacteroides and Alistipes were more frequently represented in adults, but less frequently represented in children with depression. The available data suggest some genera have a different abundance in individual psychiatric disorders (e.g., Bilophila, Bifidobacterium, Clostridium, Coprococcus, Faecalibacterium, and Ruminococcus), suggesting their importance for the gut-brain axis. Other bacterial genera might be more important for the pathophysiology of specific disorder in children and adolescents, as Akkermansia and Desulfovibrio for ASD, or Romboutsia for DD. Based on the research findings, we assume that gut microbiota corrections have the potential to improve clinical symptoms in psychiatric patients.
Collapse
Affiliation(s)
- Marcela Soltysova
- Academic Research Center for Autism, Institute of Physiology, Faculty of Medicine in Bratislava, Comenius University, 813 72 Bratislava, Slovakia
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| | - Aleksandra Tomova
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| | - Daniela Ostatnikova
- Child Psychiatry Outpatient Care Unit, Zvolen Hospital, 960 01 Zvolen, Slovakia
| |
Collapse
|
34
|
Liu Y, Chen L, Gao L, Pei X, Tao Z, Xu Y, Li R. LRRK2 deficiency protects the heart against myocardial infarction injury in mice via the P53/HMGB1 pathway. Free Radic Biol Med 2022; 191:119-127. [PMID: 36055602 DOI: 10.1016/j.freeradbiomed.2022.08.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 08/16/2022] [Accepted: 08/27/2022] [Indexed: 11/30/2022]
Abstract
LRRK2 is a Ser/Thr kinase with multiple functional domains. Studies have shown that LRRK2 mutations are closely related to hereditary Parkinson's disease. However, its role in cardiovascular disease, especially in myocardial infarction, is unclear. The aim of this study was to explore the functional role of LRRK2 in myocardial infarction. Wild-type and LRRK2-knockout mice were subjected to coronary artery ligation (left anterior descending) to establish a myocardial infarction model. Neonatal rat cardiomyocytes were subjected to hypoxia to induce hypoxic injury in vitro. We found increased LRRK2 expression levels in the infarct periphery in mouse hearts and hypoxic cardiomyocytes. LRRK2-deficient mice exhibited decreased death rates and reduced infarction areas compared to wild-type controls 14 days after infarction. LRRK2-deficient mice showed reduced left ventricular fibrosis and inflammatory responses, as well as improved cardiac function. In the in vitro study, LRRK2 silencing decreased cleaved caspase-3 activity, reduced cardiomyocyte apoptosis, and diminished hypoxia-induced inflammation. However, LRRK2 overexpression enhanced cleaved caspase-3 activity, increased the number of apoptotic cardiomyocytes, and caused remarkable hypoxia-induced inflammation. When examining the underlying mechanisms, we found that hypoxia increased HIFα expression, which enhanced LRRK2 expression. LRRK2 induced high expression of HMGB1 via P53. When HMGB1 was blocked using an anti-HMGB1 antibody, the deleterious effects caused by LRRK2 overexpression following hypoxia were inhibited in cardiomyocytes. In summary, LRRK2 deficiency protects the heart against myocardial infarction injury. The mechanism underlying this effect involves the P53-HMGB1 pathway.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lu Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Lu Gao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Xiaoxin Pei
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Zekai Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Yawei Xu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ran Li
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
35
|
Wang X, Simayi A, Fu J, Zhao X, Xu G. Resveratrol mediates the miR-149/HMGB1 axis and regulates the ferroptosis pathway to protect myocardium in endotoxemia mice. Am J Physiol Endocrinol Metab 2022; 323:E21-E32. [PMID: 35532075 DOI: 10.1152/ajpendo.00227.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endotoxemia is a common complication often used to model the acute inflammatory response associated with endotoxemia. Resveratrol has been shown to exert a wide range of therapeutic effects due to its anti-inflammatory and antioxidant properties. This study explored the effect of resveratrol on endotoxemia. Lipopolysaccharide (LPS)-induced endotoxemia mouse model and endotoxemia myocardial injury cell model were established and treated with resveratrol. Cardiomyocyte activity, lactate dehydrogenase (LDH) content in cell supernatant, glutathione (GSH) consumption, lipid reactive oxygen species (ROS) production, and iron accumulation were detected. Cardiac function indexes [left ventricular end-diastolic diameter (LVEDD), left ventricular end-systolic diameter (LVESD), ejection fraction (EF)%, and fractional shortening (FS)%] were measured using echocardiography. The creatine kinase muscle/brain isoenzyme (CK-MB) and CK levels in the serum were detected using an automatic biochemical analyzer. The downstream target of miR-149 was predicted, and the binding relationship between miR-149 and high mobility group box 1 (HMGB1) was verified using a dual-luciferase assay. miR-149 and HMGB1 expressions were detected using RT-qPCR and Western blot. After resveratrol treatment, cardiomyocyte viability and GSH were increased, and LDH secretion, lipid ROS production, lipid peroxidation, and iron accumulation were decreased, and cardiac function and cardiomyocyte injury were improved. Resveratrol improved LPS-induced endotoxemia cardiomyocyte injury by upregulating miR-149 and inhibiting ferroptosis. Resveratrol inhibited HMGB1 expression by upregulating miR-149. HMGB1 upregulation reversed the inhibitory effect of miR-149 on LPS-induced ferroptosis in cardiomyocytes. Resveratrol upregulated miR-149 and downregulated HMGB1 to inhibit ferroptosis and improve myocardial injury in mice with LPS-induced endotoxemia. Collectively, resveratrol upregulated miR-149, downregulated HMGB1, and inhibited the ferroptosis pathway, thus improving cardiomyocyte injury in LPS-induced endotoxemia.NEW & NOTEWORTHY Sepsis is an unusual systemic reaction. Resveratrol is involved in sepsis treatment. This study explored the mechanism of resveratrol in sepsis by regulating the miR-149/HMGB1 axis.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Ürümqi, People's Republic of China
| | - Alimujiang Simayi
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Ürümqi, People's Republic of China
| | - Juan Fu
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Ürümqi, People's Republic of China
| | - Xuan Zhao
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Ürümqi, People's Republic of China
| | - Guiping Xu
- Department of Anesthesiology, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Clinical Research Center for Anesthesia Management, Ürümqi, People's Republic of China
| |
Collapse
|
36
|
Transcriptomic Profiling Reveals That HMGB1 Induces Macrophage Polarization Different from Classical M1. Biomolecules 2022; 12:biom12060779. [PMID: 35740904 PMCID: PMC9221381 DOI: 10.3390/biom12060779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are key inflammatory immune cells that display dynamic phenotypes and functions in response to their local microenvironment. In different conditions, macrophage polarization can be induced by high-mobility group box 1 (HMGB1), a nuclear DNA-binding protein that activates innate immunity via the Toll-like receptor (TLR) 4, the receptor for advanced glycation end products (RAGE), and C-X-C chemokine receptor (CXCR) 4. This study investigated the phenotypes of murine bone-marrow-derived macrophages (BMDMs) stimulated with different HMGB1 redox isoforms using bulk RNA sequencing (RNA-Seq). Disulfide HMGB1 (dsHMGB1)-stimulated BMDMs showed a similar but distinct transcriptomic profile to LPS/IFNγ- and LPS-stimulated BMDMs. Fully reduced HMGB1 (frHMGB1) did not induce any significant transcriptomic change. Interestingly, compared to LPS/IFNγ- and LPS-, dsHMGB1-stimulated BMDMs showed lipid metabolism and foam cell differentiation gene set enrichment, and oil red O staining revealed that both dsHMGB1 and frHMGB1 alleviated oxidized low-density lipoprotein (oxLDL)-induced foam cells formation. Overall, this work, for the first time, used transcriptomic analysis by RNA-Seq to investigate the impact of HMGB1 stimulation on BMDM polarization. Our results demonstrated that dsHMGB1 and frHMGB1 induced distinct BMDM polarization phenotypes compared to LPS/IFNγ- and LPS- induced phenotypes.
Collapse
|
37
|
Yang X, Zhang B, Yu P, Liu M, Zhang C, Su E, Xie S, Zou Y, Jiang H, Ge J. HMGB1 in macrophage nucleus protects against pressure overload induced cardiac remodeling via regulation of macrophage differentiation and inflammatory response. Biochem Biophys Res Commun 2022; 611:91-98. [PMID: 35483224 DOI: 10.1016/j.bbrc.2022.04.053] [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: 04/01/2022] [Accepted: 04/12/2022] [Indexed: 01/03/2023]
Abstract
Pressure overload induced cardiac remodeling is associated with a complex spectrum of pathophysiological mechanisms. As inflammatory cells, macrophages maintain a critical position in mechanical stress-induced myocardial remodeling. HMGB1 is a highly conserved, ubiquitous protein in various types of cells whose biological roles are closely dependent on subcellular sites. However, whether HMGB1 expressed in macrophages performs the protective or pathological responses in cardiac remodeling is unknown. In this study, we generated the myeloid-specific HMGB1 knockout mice and detected the effects of macrophage HMGB1 in response to pathophysiological stress. Our data showed HMGB1 in macrophages played a protective role against the pressure overload induced cardiac pathophysiology. The deletion of HMGB1 in macrophages gains more differentiation of M1-type pro-inflammatory macrophage during the mechanical stress-induced myocardial remodeling, thereby aggravating the inflammatory response in whole heart, resulting in accelerated deterioration of cardiac function. Moreover, in vitro data also validated HMGB1 got involved in the process of macrophage polarization. Macrophages without HMGB1 are more inclined to differentiate into M1 during the stretch process. In summary, the present results indicated that loss of HMGB1 in macrophages can exacerbate heart failure through increased differentiation of pro-inflammatory macrophages and enhanced inflammatory response.
Collapse
Affiliation(s)
- Xue Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Baoli Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, Fudan Institute of Metabolic Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Ming Liu
- Department of Health Management Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Chunyu Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Enyong Su
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shiyao Xie
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Hong Jiang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| |
Collapse
|
38
|
Kim JO, Baek SE, Jeon EY, Choi JM, Jang EJ, Kim CD. PDGFR-β signaling mediates HMGB1 release in mechanically stressed vascular smooth muscle cells. PLoS One 2022; 17:e0265191. [PMID: 35294955 PMCID: PMC8926240 DOI: 10.1371/journal.pone.0265191] [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: 07/04/2021] [Accepted: 02/18/2022] [Indexed: 11/19/2022] Open
Abstract
Mechanically stressed vascular smooth muscle cells (VSMCs) have potential roles in the development of vascular complications. However, the underlying mechanisms are unclear. Using VSMCs cultured from rat thoracic aorta explants, we investigated the effects of mechanical stretch (MS) on the cellular secretion of high mobility group box 1 (HMGB1), a major damage-associated molecular pattern that mediates vascular complications in stressed vasculature. Enzyme-linked immunosorbent assay (ELISA) demonstrated an increase in the secretion of HMGB1 in VSMCs stimulated with MS (0–3% strain, 60 cycles/min), and this secretion was markedly and time-dependently increased at 3% MS. The increased secretion of HMGB1 at 3% MS was accompanied by an increased cytosolic translocation of nuclear HMGB1; the acetylated and phosphorylated forms of this protein were significantly increased. Among various inhibitors of membrane receptors mediating mechanical signals, AG1295 (a platelet-derived growth factor receptor (PDGFR) inhibitor) attenuated MS-induced HMGB1 secretion. Inhibitors of other receptors, including epidermal growth factor, insulin-like growth factor, and fibroblast growth factor receptors, did not inhibit this secretion. Additionally, MS-induced HMGB1 secretion was markedly attenuated in PDGFR-β-deficient cells but not in cells transfected with PDGFR-α siRNA. Likewise, PDGF-DD, but not PDGF-AA, directly increased HMGB1 secretion in VSMCs, indicating a pivotal role of PDGFR-β signaling in the secretion of this protein in VSMCs. Thus, targeting PDGFR-β-mediated secretion of HMGB1 in VSMCs might be a promising therapeutic strategy for vascular complications associated with hypertension.
Collapse
Affiliation(s)
- Ji On Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Seung Eun Baek
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Eun Yeong Jeon
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Jong Min Choi
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Eun Jeong Jang
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
| | - Chi Dae Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Yangsan, Gyeongnam, Republic of Korea
- * E-mail:
| |
Collapse
|
39
|
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: 6.3] [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
|
40
|
Xie SS, Deng Y, Guo SL, Li JQ, Zhou YC, Liao J, Wu DD, Lan WF. Endothelial cell ferroptosis mediates monocrotaline-induced pulmonary hypertension in rats by modulating NLRP3 inflammasome activation. Sci Rep 2022; 12:3056. [PMID: 35197507 PMCID: PMC8866506 DOI: 10.1038/s41598-022-06848-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 01/28/2022] [Indexed: 12/18/2022] Open
Abstract
Inflammation triggers pulmonary vascular remodelling. Ferroptosis, a nonapoptotic form of cell death that is triggered by iron-dependent lipid peroxidation and contributes to the pathogenesis of several inflammation-related diseases, but its role in pulmonary hypertension (PH) has not been studied. We examined endothelial cell ferroptosis in PH and the potential mechanisms. Pulmonary artery endothelial cells (PAECs) and lung tissues from monocrotaline (MCT)-induced PH rats were analysed for ferroptosis markers, including lipid peroxidation, the labile iron pool (LIP) and the protein expression of glutathione peroxidase 4 (GPX4), ferritin heavy chain 1 (FTH1) and NADPH oxidase-4 (NOX4). The effects of the ferroptosis inhibitor ferrostatin-1 (Fer-1) on endothelial cell ferroptosis and pulmonary vascular remodelling in MCT-induced rats were studied in vitro and in vivo. Ferroptosis was observed in PAECs from MCT-induced PH rats in vitro and in vivo and was characterized by a decline in cell viability accompanied by increases in the LIP and lipid peroxidation, the downregulation of GPX4 and FTH1 expression and the upregulation of NOX4 expression. High-mobility group box 1 (HMGB1)/Toll-like receptor 4 (TLR4)/NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome signalling was measured by western blotting. These changes were significantly blocked by Fer-1 administration in vitro and in vivo. These results suggest that Fer-1 plays a role in inhibiting ferroptosis-mediated PAEC loss during the progression of PH. The ferroptosis-induced inflammatory response depended on the activation of HMGB1/TLR4 signalling, which activated the NLRP3 inflammasome in vivo. We are the first to suggest that pulmonary artery endothelial ferroptosis triggers inflammatory responses via the HMGB1/TLR4/NLRP3 inflammasome signalling pathway in MCT-induced rats. Treating PH with a ferroptosis inhibitor and exploring new treatments based on ferroptosis regulation might be promising therapeutic strategies for PH.
Collapse
Affiliation(s)
- Shan-Shan Xie
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China
| | - Yan Deng
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China.
- Department of Echocardiography of Cardiovascular Disease Institute, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China.
| | - Sheng-Lan Guo
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China
| | - Jia-Quan Li
- Experimental Centre of Guangxi Medical University, Nanning, People's Republic of China
| | - Ying-Chuan Zhou
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
| | - Juan Liao
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China
| | - Dan-Dan Wu
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China
| | - Wei-Fang Lan
- Department of Ultrasound, First Affiliated Hospital of Guangxi Medical University, 6 Shuang yong Road, Nanning, 530021, People's Republic of China
| |
Collapse
|
41
|
Zhang X, Wang T, Chen Z, Wang H, Yin Y, Wang L, Wang Y, Xu B, Xu W. HMGB1‐Promoted Neutrophil Extracellular Traps Contribute to Cardiac Diastolic Dysfunction in Mice. J Am Heart Assoc 2022; 11:e023800. [PMID: 35156391 PMCID: PMC9245819 DOI: 10.1161/jaha.121.023800] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background Heart failure with preserved ejection fraction (HFpEF) remains an increasing public health problem with substantial morbidity and mortality but with few effective treatments. A novel inflammatory mechanism has been proposed, but the inflammatory signals promoting the development of HFpEF remain greatly unknown. Methods and Results Serum of patients with HFpEF was collected for measurement of circulating neutrophils and markers of neutrophil extracellular traps (NETs). To induce HFpEF phenotype, male C57BL/6 mice underwent uninephrectomy, received a continuous infusion of d‐aldosterone for 4 weeks, and maintained on 1.0% sodium chloride drinking water. Heart tissues were harvested, immune cell types determined by flow cytometry, NETs formation by immunofluorescence, and western blotting. Differentiated neutrophils were cultured to investigate the effect of HMGB1 (high mobility group protein B1) and SGLT2 (sodium‐glucose cotransporter‐2) inhibitor on NETs formation in vitro. Circulating neutrophils and NETs markers are elevated in patients with HFpEF, as are cardiac neutrophils and NETs formation in HFpEF mice. NETs inhibition with deoxyribonuclease 1 in experimental HFpEF mice reduces heart macrophages infiltration and inflammation and ameliorates cardiac fibrosis and diastolic function. Damage‐associated molecular pattern HMGB1 expression is elevated in cardiac tissue of HFpEF mice, and HMGB1 inhibition reduces heart neutrophil infiltration and NETs formation and ameliorates diastolic function. Lastly, SGLT2 inhibitor empagliflozin down‐regulates heart HMGB1 expression, attenuates NETs formation and cardiac fibrosis, and improves diastolic function in HFpEF mice. Conclusions NETs contribute to the pathogenesis of HFpEF, which can be ameliorated by HMGB1 inhibition and SGLT2 inhibitors. Thus, HMGB1 and NETs may represent novel therapeutic targets for the treatment of HFpEF.
Collapse
Affiliation(s)
- Xin‐Lin Zhang
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| | - Ting‐Yu Wang
- Central for Translational Medicine Nanjing University School of Medicine Nanjing China
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine Nanjing University School of Medicine Nanjing China
| | - Zheng Chen
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| | - Hong‐Wei Wang
- Central for Translational Medicine Nanjing University School of Medicine Nanjing China
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine Nanjing University School of Medicine Nanjing China
| | - Yong Yin
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| | - Lian Wang
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| | - Yong Wang
- Central for Translational Medicine Nanjing University School of Medicine Nanjing China
- State Key Laboratory of Analytical Chemistry for Life Science & Jiangsu Key Laboratory of Molecular Medicine Nanjing University School of Medicine Nanjing China
| | - Biao Xu
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| | - Wei Xu
- Department of Cardiology Affiliated Drum Tower Hospital Nanjing University School of Medicine Nanjing China
| |
Collapse
|
42
|
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: 402] [Impact Index Per Article: 134.0] [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
|
43
|
Foglio E, Pellegrini L, Russo MA, Limana F. HMGB1-Mediated Activation of the Inflammatory-Reparative Response Following Myocardial Infarction. Cells 2022; 11:cells11020216. [PMID: 35053332 PMCID: PMC8773872 DOI: 10.3390/cells11020216] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/04/2022] [Accepted: 01/08/2022] [Indexed: 02/06/2023] Open
Abstract
Different cell types belonging to the innate and adaptive immune system play mutually non-exclusive roles during the different phases of the inflammatory-reparative response that occurs following myocardial infarction. A timely and finely regulation of their action is fundamental for the process to properly proceed. The high-mobility group box 1 (HMGB1), a highly conserved nuclear protein that in the extracellular space can act as a damage-associated molecular pattern (DAMP) involved in a large variety of different processes, such as inflammation, migration, invasion, proliferation, differentiation, and tissue regeneration, has recently emerged as a possible regulator of the activity of different immune cell types in the distinct phases of the inflammatory reparative process. Moreover, by activating endogenous stem cells, inducing endothelial cells, and by modulating cardiac fibroblast activity, HMGB1 could represent a master regulator of the inflammatory and reparative responses following MI. In this review, we will provide an overview of cellular effectors involved in these processes and how HMGB1 intervenes in regulating each of them. Moreover, we will summarize HMGB1 roles in regulating other cell types that are involved in the different phases of the inflammatory-reparative response, discussing how its redox status could affect its activity.
Collapse
Affiliation(s)
- Eleonora Foglio
- Technoscience, Parco Scientifico e Tecnologico Pontino, 04100 Latina, Italy;
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Laura Pellegrini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Matteo Antonio Russo
- IRCCS San Raffaele Roma and MEBIC Consortium, 00166 Rome, Italy;
- San Raffaele University of Rome, 00166 Rome, Italy
| | - Federica Limana
- San Raffaele University of Rome, 00166 Rome, Italy
- Laboratory of Cellular and Molecular Pathology, IRCCS San Raffaele Roma, 00166 Rome, Italy
- Correspondence:
| |
Collapse
|
44
|
Wang W, Zheng F, Zhang A. Arsenic-induced lung inflammation and fibrosis in a rat model: Contribution of the HMGB1/RAGE, PI3K/AKT, and TGF-β1/SMAD pathways. Toxicol Appl Pharmacol 2021; 432:115757. [PMID: 34673086 DOI: 10.1016/j.taap.2021.115757] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023]
Abstract
An increasing number of studies have shown that arsenic exposure increases the risk of lung cancer as well as a variety of non-malignant respiratory diseases, including bronchitis and tracheobronchitis. HMGB1 is widely expressed in a variety of tissues and cells and is involved in the pathological processes of many lung diseases through binding to the corresponding receptors and activating the downstream signaling pathways. However, the exact role of HMGB1/RAGE in arsenic-induced lung injury remains unknown. The aim of this study was to investigate whether HMGB1/RAGE and its activated downstream pathways are involved in the process of arsenic exposure-induced lung injury in rats. In this study, an animal model of oral exposure to arsenic was induced using 2.5, 5 and 10 mg/kg NaAsO2. The results showed that capillary permeability (LDH, TP, ACP, and AKP) was increased in the arsenic exposure groups, resulting in cell damage; this was accompanied by acute inflammation marked by significant neutrophil infiltration. Meanwhile, obvious histopathological damage, including thickening of the lung epithelium, increased infiltration of inflammatory cells, rupture of the alveolar wall, swelling of the mitochondria, and chromatin agglutination was observed by H&E staining and transmission electron microscopy. Furthermore, the results confirmed that the expressions of HMGB1 and RAGE in lung tissue were enhanced, and protein expression of PI3K, p-AKT, IL-1β, IL-18, and MMP-9 was increased in lung homogenates from the arsenic-exposed groups compared to the control group. Finally, Masson's staining results revealed arsenic-induced fibrosis and collagen deposition. Moreover, a significant increase in key fibrosis factors, including TGF-β1, p-SMAD2, p-SMAD3, and SMAD4 was observed in the lung homogenates in arsenic-exposed groups. In conclusion, the current study demonstrates that sub-chronic arsenic exposure triggers the inflammatory response and collagen fiber deposition in rat lung tissue. The potential mechanism may be closely related to activation of the pro-inflammatory-related HMGB1/RAGE pathway and initiation of the PI3K/AKT and TGF-β1/SMAD pathways.
Collapse
Affiliation(s)
- Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Fanyan Zheng
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, Guizhou, PR China.
| |
Collapse
|
45
|
Bayer AL, Alcaide P. MyD88: At the heart of inflammatory signaling and cardiovascular disease. J Mol Cell Cardiol 2021; 161:75-85. [PMID: 34371036 PMCID: PMC8629847 DOI: 10.1016/j.yjmcc.2021.08.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/27/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease is a leading cause of death worldwide and is associated with systemic inflammation. In depth study of the cell-specific signaling mechanisms mediating the inflammatory response is vital to improving anti-inflammatory therapies that reduce mortality and morbidity. Cellular damage in the cardiovascular system results in the release of damage associated molecular patterns (DAMPs), also known as "alarmins," which activate myeloid cells through the adaptor protein myeloid differentiation primary response 88 (MyD88). MyD88 is broadly expressed in most cell types of the immune and cardiovascular systems, and its role often differs in a cardiovascular disease context and cell specific manner. Herein we review what is known about MyD88 in the setting of a variety of cardiovascular diseases, discussing cell specific functions and the relative contributions of MyD88-dependent vs. independent alarmin triggered inflammatory signaling. The widespread involvement of these pathways in cardiovascular disease, and their largely unexplored complexity, sets the stage for future in depth mechanistic studies that may place MyD88 in both immune and non-immune cell types as an attractive target for therapeutic intervention in cardiovascular disease.
Collapse
Affiliation(s)
- Abraham L Bayer
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine. 136 Harrison Ave, Boston, MA 02111, United States of America.
| |
Collapse
|
46
|
High mobility group box 1 and homocysteine as preprocedural predictors for contrast-induced acute kidney injury after percutaneous coronary artery intervention. Int Urol Nephrol 2021; 54:1663-1671. [PMID: 34727314 DOI: 10.1007/s11255-021-03050-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 10/24/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE High mobility group box 1 (HMGB1) and homocysteine (Hcy) play important roles in contrast-induced acute kidney injury (CI-AKI). We compared HMGB1 to Hcy as preprocedural predictors for CI-AKI in coronary artery disease (CAD) patients after percutaneous coronary artery intervention (PCI). METHODS We included 257 eligible patients who were categorized into CI-AKI ( +) and CI-AKI ( -) group. The differences in clinical characteristics and biochemical indexes between two groups were analyzed. RESULTS We observed that thirty-eight (14.8%) of 257 eligible CAD patients developed CI-AKI. HMGB1 (14.65 [11.13-24.89] vs 10.88 [7.94-13.23], p < 0.001) and Hcy (14.07 [12.07-17.31] vs 12.09 [10.71-13.47], p < 0.001) increased significantly in CI-AKI ( +) group. Both age (r = 0.210, p = 0.001), serum creatinine (r = 0.509, p < 0.001), eGFR (r = - 0.459, p < 0.001) and Hcy (r = 0.531, p < 0.001) were significantly correlated with HMGB1. Among all patients, HMGB1 (OR 1.181, 95% CI 1.081-1.290, p < 0.001) and Hcy (OR 1.260, 95% CI 1.066-1.489, p = 0.007) were independent predictors for the development of CI-AKI. We built the propensity score matching (PSM) using 38 pairs of patients. After adjustment, HMGB1 (OR 1.169, 95% CI 1.035-1.322, p = 0.012) and Hcy (OR 1.457, 95% CI 1.064-1.997, p = 0.019) were also independent predictors for the development of CI-AKI. Both HMGB1 (AUC: 0.704, 95% CI: 0.588-0.819, p = 0.002) and Hcy (AUC: 0.708, 95% CI: 0.593-0.823, p = 0.002) had predictive values for CI-AKI. CONCLUSION There is a significant positive association between HMGB1 and Hcy in CAD patients. Both HMGB1 and Hcy are potential preprocedural predictors of CI-AKI after PCI.
Collapse
|
47
|
Yi C, Zhang X, Li H, Chen G, Zeng B, Li Y, Wang C, He Y, Chen X, Huang Z, Yu D. EPHB4 Regulates the Proliferation and Metastasis of Oral Squamous Cell Carcinoma through the HMGB1/NF-κB Signalling Pathway. J Cancer 2021; 12:5999-6011. [PMID: 34539874 PMCID: PMC8425198 DOI: 10.7150/jca.59331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 08/08/2021] [Indexed: 11/05/2022] Open
Abstract
Background: Malignant proliferation and cervical lymphatic metastasis restrict the prognosis of oral squamous cell carcinoma (OSCC). Erythropoietin-producing human hepatocellular B4 (EPHB4) regulates a series of tumour functions involving tumourigenesis, cancer cell attachment and metastasis. However, the mechanism of EphB4 regulating the malignant progression of OSCC has not been fully elucidated. Methods: EPHB4 expression was analysed in 65 OSCC samples and adjacent noncancerous tissues through immunohistochemistry (IHC). siRNA and overexpression plasmids were transfected into OSCC cells to modify EPHB4 expression, and then, regulatory functions were explored in vitro and in vivo. Co-immunoprecipitation (Co-IP) and mass spectrometry were applied to detect proteins interacting with EPHB4. Subsequently, protein stability assays and NF-κB pathway inhibition assays were used to verify the regulation of EPHB4, high-mobility group box 1 (HMGB1) and nuclear factor-κB (NF-κB) activation. Results: EPHB4 was found to be highly expressed in OSCC tissues, which was related to tumour stage and lymphatic metastasis and resulted in a poor prognosis. Cellular experiments and mouse tongue xenograft models further confirmed that high EPHB4 expression promoted the proliferation and metastasis of OSCC tumours. Mechanistically, co-IP and mass spectrometry studies indicated that EPHB4 could bind to HMGB1 and maintain HMGB1 stability. Downregulation of HMGB1 inhibited the proliferation and metastasis of OSCC cells and inhibited NF-κB phosphorylation activation but did not affect EPHB4 expression. Conclusion: This study revealed the mechanism by which EPHB4 promotes the proliferation and metastasis of OSCC by activating the HMGB1-mediated NF-κB signalling pathway, which can be exploited as a novel marker or therapeutic target to control metastasis and improve the survival rate of OSCC.
Collapse
Affiliation(s)
- Chen Yi
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Xiliu Zhang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Hongyu Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Guanhui Chen
- Department of Stomatology, the Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen. Guangdong, China, 518107
| | - Binghui Zeng
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Yiming Li
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Chao Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Yi He
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Xun Chen
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| | - Zixian Huang
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China, 510120
| | - Dongsheng Yu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University. Guangzhou, Guangdong, China, 510055
- Guangdong Provincial Key Laboratory of Stomatology. Guangzhou, Guangdong, China, 510055
| |
Collapse
|
48
|
Recombinant High-Mobility Group Box 1 (rHMGB1) Promotes NRF2-Independent Mitochondrial Fusion through CXCR4/PSMB5-Mediated Drp1 Degradation in Endothelial Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:9993240. [PMID: 34394840 PMCID: PMC8358426 DOI: 10.1155/2021/9993240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/22/2021] [Accepted: 07/08/2021] [Indexed: 11/17/2022]
Abstract
Mitochondrial dynamics plays an important role in maintaining normal endothelial cell function and in the pathogenesis of cardiovascular disease. It is not identified whether high-mobility group box 1 (HMGB1), a representative damage-associated molecular pattern (DAMP) molecule, could influence mitochondrial dynamics in endothelial cells. The objective of this study is to clarify the effect of HMGB1 on mitochondrial dynamics in endothelial cells and the underlying mechanism. EA.hy926 human endothelial cells were incubated with recombinant HMGB1 (rHMGB1); mitochondrial morphology was observed with a confocal microscope and transmission electron microscope (TEM). The expression of dynamin-related protein 1 (Drp1), Mitofusin 1 (Mfn1), Mitofusin 2 (Mfn2), Optic atrophy 1 (Opa1), phosphatase and tensin homolog- (PTEN-) induced kinase 1 (PINK1), NOD-like receptor 3 (NLRP3), caspase 1, cleaved caspase 1, 20S proteasome subunit beta 5 (PSMB5), and antioxidative master nuclear factor E2-related factor 2 (NRF2) and the concentration of interleukin 1β (IL-1β) were determined. Specific inhibitors C29, TAK-242, FPS-ZM1, AMD3100, and epoxomicin were used to block toll-like receptor 2 (TLR2), toll-like receptor 4 (TLR4), receptor for advanced glycation end products (RAGE), C-X-C-chemokine receptor 4 (CXCR4), and PSMB5, respectively. siRNAs were used to silence the expression of NRF2. rHMGB1 promoted mitochondrial fusion in endothelial cells, while no significant proinflammatory effects were found. The expression of mitochondrial fission protein Drp1 and phosphorylated subtypes p-Drp1-S616 and p-Drp1-S637 were all downregulated; no significant expression changes of PINK1 and Mfn1, Mfn2, and Opa1 were found. Inhibition of CXCR4 but not TLR4, RAGE, or TLR2 reversed rHMGB1-induced Drp1 downregulation and mitochondrial fusion. Interestingly, inhibition of TLR4 with TAK-242 promoted Drp1 downregulation and mitochondrial fusion. rHMGB1 increased the expression of NRF2 and PSMB5; inhibition of PSMB5 but not silencing NRF2 abolished rHMGB1-induced Drp1 downregulation and mitochondrial fusion. These results indicate that rHMGB1 promotes NRF2 independent mitochondrial fusion via CXCR4/PSMB5 pathway-mediated Drp1 proteolysis. rHMGB1 may influence mitochondrial and endothelial function through this effect on mitochondrial dynamics.
Collapse
|
49
|
Wu J, Wu Z, He A, Zhang T, Zhang P, Jin J, Li S, Li G, Li X, Liang S, Pei L, Liu R, Tian Q, He X, Lu Y, Tang Z, Li H. Genome-Wide Screen and Validation of Microglia Pro-Inflammatory Mediators in Stroke. Aging Dis 2021; 12:786-800. [PMID: 34094642 PMCID: PMC8139211 DOI: 10.14336/ad.2020.0926] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022] Open
Abstract
Stroke activates microglia pro-inflammatory response that not only induces the early neuronal injuries but also causes the secondary brain infarction. Yet, the underlying mechanisms for how microglia become activated in stroke are still unknown. Here, using the next-generation of RNA sequencing we find a total of 778 genes increasingly expressed in brain of stroke mice. Of these, we identified Hmgb2 as a microglia pro-inflammatory mediator by promoting the transcription of Ctss. Inhibition of either Hmgb2 or Ctss blocks microglia pro-inflammatory response and protects against brain damages and improves the neurological functions of stroke mice. This study uncovers Hmgb2 and Ctss as the major microglia inflammatory response mediators in stroke and hence warrants the promising targets for stroke therapies.
Collapse
Affiliation(s)
- Jianhua Wu
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,3Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhuoze Wu
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Aodi He
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Tongmei Zhang
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ping Zhang
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,4Department of Neurology, Tongji Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Jin
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Sisi Li
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Gaigai Li
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,4Department of Neurology, Tongji Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyan Li
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Shiqi Liang
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Pei
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,5Department of Neurobiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Liu
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,6Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Tian
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,6Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ximiao He
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youming Lu
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Zhouping Tang
- 2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China.,4Department of Neurology, Tongji Hospital, Tongji College of Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Li
- 1Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
50
|
Zhang C, Liang Z, Ma S, Liu X. Radiotherapy and Cytokine Storm: Risk and Mechanism. Front Oncol 2021; 11:670464. [PMID: 34094967 PMCID: PMC8173139 DOI: 10.3389/fonc.2021.670464] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 04/20/2021] [Indexed: 12/27/2022] Open
Abstract
Radiotherapy (RT) shows advantages as one of the most important precise therapy strategies for cancer treatment, especially high-dose hypofractionated RT which is widely used in clinical applications due to the protection of local anatomical structure and relatively mild impairment. With the increase of single dose, ranging from 2~20 Gy, and the decrease of fractionation, the question that if there is any uniform standard of dose limits for different therapeutic regimens attracts more and more attention, and the potential adverse effects of higher dose radiation have not been elucidated. In this study, the immunological adverse responses induced by radiation, especially the cytokine storm and the underlying mechanisms such as DAMPs release, pro-inflammatory cytokine secretion and cGAS-STING pathway activation, will be elucidated, which contributes to achieving optimal hypofractionated RT regimen, improving the killing of cancer cells and avoiding the severe side effects.
Collapse
Affiliation(s)
- Chen Zhang
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zhenzhen Liang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Shumei Ma
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Liu
- School of Public Health and Management, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|