1
|
Li J, Wang Z, Li J, Zhao H, Ma Q. HMGB1: A New Target for Ischemic Stroke and Hemorrhagic Transformation. Transl Stroke Res 2025; 16:990-1015. [PMID: 38740617 PMCID: PMC12045843 DOI: 10.1007/s12975-024-01258-5] [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: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 05/16/2024]
Abstract
Stroke in China is distinguished by its high rates of morbidity, recurrence, disability, and mortality. The ultra-early administration of rtPA is essential for restoring perfusion in acute ischemic stroke, though it concurrently elevates the risk of hemorrhagic transformation. High-mobility group box 1 (HMGB1) emerges as a pivotal player in neuroinflammation after brain ischemia and ischemia-reperfusion. Released passively by necrotic cells and actively secreted, including direct secretion of HMGB1 into the extracellular space and packaging of HMGB1 into intracellular vesicles by immune cells, glial cells, platelets, and endothelial cells, HMGB1 represents a prototypical damage-associated molecular pattern (DAMP). It is intricately involved in the pathogenesis of atherosclerosis, thromboembolism, and detrimental inflammation during the early phases of ischemic stroke. Moreover, HMGB1 significantly contributes to neurovascular remodeling and functional recovery in later stages. Significantly, HMGB1 mediates hemorrhagic transformation by facilitating neuroinflammation, directly compromising the integrity of the blood-brain barrier, and enhancing MMP9 secretion through its interaction with rtPA. As a systemic inflammatory factor, HMGB1 is also implicated in post-stroke depression and an elevated risk of stroke-associated pneumonia. The role of HMGB1 extends to influencing the pathogenesis of ischemia by polarizing various subtypes of immune and glial cells. This includes mediating excitotoxicity due to excitatory amino acids, autophagy, MMP9 release, NET formation, and autocrine trophic pathways. Given its multifaceted role, HMGB1 is recognized as a crucial therapeutic target and prognostic marker for ischemic stroke and hemorrhagic transformation. In this review, we summarize the structure and redox properties, secretion and pathways, regulation of immune cell activity, the role of pathophysiological mechanisms in stroke, and hemorrhage transformation for HMGB1, which will pave the way for developing new neuroprotective drugs, reduction of post-stroke neuroinflammation, and expansion of thrombolysis time window.
Collapse
Affiliation(s)
- Jiamin Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Zixin Wang
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Jiameng Li
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China
| | - Haiping Zhao
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| | - Qingfeng Ma
- Department of Neurology and Cerebrovascular Diseases Research Institute, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Beijing, China.
| |
Collapse
|
2
|
Han JY, Rhee WJ, Shin JS. Cytoplasmic HMGB1 promotes the activation of JAK2-STAT3 signaling and PD-L1 expression in breast cancer. Mol Med 2025; 31:197. [PMID: 40389855 PMCID: PMC12090602 DOI: 10.1186/s10020-025-01235-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/28/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND High-mobility group box 1 (HMGB1) plays various roles depending on its subcellular localization. Extracellular HMGB1 interacts with receptors, such as toll-like receptor 4 and receptor for advanced glycation end products (RAGE), promoting cell proliferation, survival, and migration in cancer cells. It also increases the expression of programmed death-ligand 1 (PD-L1) in cancer cells by binding to RAGE. However, the effect of intracellular HMGB1 on the regulation of immune checkpoints such as PD-L1 has not been well characterized. In this study, we aimed to investigate the effects of intracellular HMGB1 on PD-L1 expression in breast cancer cells. METHODS Human and mouse triple-negative breast cancer cells, MDA-MB-231 and 4T1, along with HMGB1-deficient mouse embryonic fibroblast cells, were cultured. HMGB1 overexpression was achieved using a Myc-tagged plasmid, while siHMGB1 constructs were used for gene silencing. Quantitative reverse-transcriptase PCR and western blot analysis were performed to assess gene and protein expressions. Confocal imaging, immunoprecipitation, and proximity ligation assays were used to investigate HMGB1 localization and Janus kinase 2 (JAK2)-signal transducer and activator of transcription 3 (STAT3) interactions. In vivo experiments were performed using tumor-bearing mice treated with STAT3 and HMGB1 inhibitors. Statistical analyses were performed using Student's t-tests, one-way analysis of variance, Pearson's correlation, and Kaplan-Meier survival analysis, with significance set at p < 0.05. RESULTS In breast cancer cells, HMGB1 translocation from the nucleus to the cytoplasm increased the JAK2-STAT3 interaction and induced STAT3 phosphorylation, leading to increased STAT3 target signaling, including the epithelial-mesenchymal transition (EMT) phenotype and PD-L1 expression. Inhibition of nucleo-cytoplasmic translocation of HMGB1 decreased STAT3 phosphorylation and PD-L1 expression. Furthermore, HMGB1 enhanced breast cancer cell migration, invasion, and EMT, contributing to tumor growth in an in vivo mouse model that were mitigated by the HMGB1-targeted approach. CONCLUSIONS These findings underscore the critical role of intracellular HMGB1 in modulating PD-L1 expression via the JAK2-STAT3 signaling pathways in breast cancer and suggest that targeting HMGB1 translocation is a promising strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Ju-Young Han
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Woo Joong Rhee
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, 50-1 Yonsei-ro Seodaemun-gu, Seoul, 03722, South Korea.
- Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
3
|
Wang Z, Guo W, Zhang X, Wei Y, Zhang W, Du N, Li C, Wu X, Yi F, Zhou T, Dong X, Guo Q, Xu H, Wang E, Li N, Cheng R, Li Z, Song X, Sun Y, Sun X, Cao L. Tumor microenvironment-associated oxidative stress impairs SIRT1 secretion to suppress anti-tumor immune response. Cell Rep 2025; 44:115679. [PMID: 40343797 DOI: 10.1016/j.celrep.2025.115679] [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: 05/15/2024] [Revised: 11/10/2024] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Sirtuin-1 (SIRT1) is a classical histone deacetylase well known for its roles in intracellular pathways such as energy metabolism, DNA damage response, and genome stability maintenance. We report that SIRT1 can be secreted into the tumor microenvironment (TME) through an unconventional protein secretion pathway, effectively inhibiting tumor growth. However, under the stressful conditions of the TME, SIRT1 undergoes increased methylation, which impedes its secretion. Consequently, tumor-infiltrating M2 macrophages are unable to acquire sufficient SIRT1 from the TME, resulting in a significant decrease in SIRT1 levels within these cells. This SIRT1 decline leads to elevated expression of programmed cell death ligand 1 (PD-L1) on M2 macrophages, which in turn contributes to CD8+ T cell exhaustion through the programmed cell death protein 1/PD-L1 interaction pathway. These findings unveil the multifaceted roles and regulatory mechanisms of SIRT1 within the complex TME, providing deeper insights that significantly enhance our understanding of tumor immune-evasion strategies.
Collapse
Affiliation(s)
- Zhuo Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Wendong Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaowen Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Yufei Wei
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Wanying Zhang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ning Du
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Chunlu Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xuan Wu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Fei Yi
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Tingting Zhou
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiang Dong
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China
| | - Qiqiang Guo
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Hongde Xu
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Erli Wang
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Na Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Rong Cheng
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Ziwei Li
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China
| | - Xiaoyu Song
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| | - Yingxian Sun
- Department of Cardiology, First Hospital of China Medical University, Shenyang, Liaoning 110122, China.
| | - Xun Sun
- Department of Immunology, Basic Medicine College, China Medical University, Shenyang, Liaoning 110122, China.
| | - Liu Cao
- Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, Liaoning 110122, China; Health Sciences Institute, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
4
|
Saha A, Islam MM, Kumar R, Ismail AM, Garcia E, Gullapali RR, Chodosh J, Rajaiya J. Virus and cell specific HMGB1 secretion and subepithelial infiltrate formation in adenovirus keratitis. PLoS Pathog 2025; 21:e1013184. [PMID: 40367285 PMCID: PMC12101768 DOI: 10.1371/journal.ppat.1013184] [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: 01/30/2025] [Revised: 05/23/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025] Open
Abstract
A highly contagious infection caused by human adenovirus species D (HAdV-D), epidemic keratoconjunctivitis (EKC) results in corneal subepithelial infiltration (SEI) by leukocytes, the hallmark of the infection. To date, the pathogenesis of corneal SEI formation in EKC is unresolved. HMGB1 (high-mobility group box 1 protein) is an alarmin expressed in response to infection and a marker of sepsis. Earlier studies using a different adenovirus species, HAdV-C, showed retention of HMGB1 in the infected cell nucleus by adenovirus protein VII, enabling immune evasion. Here, using HAdV-D we show cell-specific HMGB1 secretion by infected cells, and provide an HAdV-D specific mechanism for SEI formation in EKC. HMGB1 was secreted only upon infection of human corneal epithelial cells, not from other cell types, and only upon infection by HAdV-D types associated with EKC. Acetylated HMGB1 translocation from the nucleus to the cytoplasm, then to the extracellular milieu, was tightly controlled by CRM1 and LAMP1, respectively. Primary stromal cells when stimulated by rHMGB1 expressed proinflammatory chemokines. In a novel 3D culture system in tune with the architecture of the cornea, HMGB1 released by infected corneal epithelial cells induced leukocytic infiltrates either directly and/or indirectly via stimulated stromal cells, which together explains SEI formation in EKC.
Collapse
Affiliation(s)
- Amrita Saha
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Mohammad Mirazul Islam
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology and Visual Sciences, University of Ophthalmology and Visual Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Rahul Kumar
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Ashrafali Mohamed Ismail
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emanuel Garcia
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Rama R. Gullapali
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - James Chodosh
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
- Department of Ophthalmology and Visual Sciences, University of Ophthalmology and Visual Sciences, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| | - Jaya Rajaiya
- Department of Ophthalmology, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Molecular Genetics and Microbiology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, United States of America
| |
Collapse
|
5
|
Tsubota M, Sasaki K, Shin E, Okamura Y, Nishimura A, Yamagata A, Nonaka Y, Sekiguchi F, Tomono Y, Nishibori M, Okada T, Toyooka N, Kawabata A. HMGB1 derived from macrophages and enteric glial cells contributes to the butyrate-induced colonic hypersensitivity in mice. Eur J Pharmacol 2025; 999:177660. [PMID: 40274181 DOI: 10.1016/j.ejphar.2025.177660] [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: 10/31/2024] [Revised: 03/30/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025]
Abstract
High mobility group box1 (HMGB1), a nuclear protein, once acetylated by histone acetyltransferase, is released into the extracellular space, and causes pain signals, thereby contributing to pathological pain. Repeated intracolonic administration of butyrate, known to inhibit histone deacetylase (HDAC), produces colonic hypersensitivity in rodents, being widely used as models for irritable bowel syndrome (IBS). Thus, we asked whether HMGB1 would participate in the butyrate-induced colonic hypersensitivity in mice, and analyzed the underlying mechanisms. Repeated butyrate treatment caused colonic hypersensitivity to distension and intraluminal sulfide, a functional enhancer of Cav3.2 channels, in mice, which was prevented by repeated treatment with an anti-HMGB1-neutralizing antibody, thrombomodulin alfa (TMα) capable of causing thrombin-dependent degradation of HMGB1, antagonists for RAGE, TLR4 and CXCR4, membrane receptors of HMGB1, liposomal clodronate, a macrophage depletor, and ethyl pyruvate capable of inhibiting HMGB1 release from macrophages. Butyrate treatment increased the number of Iba1-positive macrophages, but not S100B-positive enteric glial cells (EGCs), and the rate of cytosolic/whole cell HMGB1 levels in both types of cells in the colonic mucosa. In macrophage-like RAW264.7 cells and EGC-like CRL-2690 cells, butyrate as well as trichostatin A, a well-known HDAC inhibitor, at the same concentrations that increased histone acetylation, evoked cytoplasmic translocation and extracellular release of nuclear HMGB1. Together, butyrate is considered to cause HMGB1 release from macrophages and EGCs most probably by inhibiting HDAC, resulting in colonic hypersensitivity in mice. HMGB1 and its membrane receptors might serve as drug targets for colonic hypersensitivity in IBS patients.
Collapse
Affiliation(s)
- Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Kana Sasaki
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Eunkyung Shin
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yuta Okamura
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Ayaka Nishimura
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Ayumu Yamagata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yui Nonaka
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan
| | - Yasuko Tomono
- Department of Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Masahiro Nishibori
- Department of Translational Research and Drug Development, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, 700-8558, Japan
| | - Takuya Okada
- Faculty of Engineering, University of Toyama, 3190 Gofuku, Toyama, 930-8555, Japan
| | - Naoki Toyooka
- Faculty of Engineering, University of Toyama, 3190 Gofuku, Toyama, 930-8555, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University, 3-4-1 Kowakae, Higashi-Osaka, 577-8502, Japan.
| |
Collapse
|
6
|
Zhang M, Ma Z, Cui H, Miao Y, Yin Y, Wen Q, Liu Z, Huang X, Xing C, Liu K, Peng H, Song L. Involvement of circadian clock protein PER2 in controlling sleep deprivation induced HMGB1 up-regulation by targeting p300 in the cortex. Sci Rep 2025; 15:12253. [PMID: 40210902 PMCID: PMC11985928 DOI: 10.1038/s41598-025-96931-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Lack of sleep is a common problem in current society, which can induce various brain dysfunctions. Neuroinflammation is a typical reaction caused by sleep deficit and is considered as a common basis for various neurological disorders and cognitive impairments, but the related mechanisms have not been fully clarified. The circadian clock protein plays a critical role in maintaining physiological homeostasis, including sleep/wake cycles. Circadian disorders induced by sleep deficit might contribute to the development of neuroinflammation. In the current study, we observed that sleep deprivation (SD) induced elevated expression of High-mobility group box 1 (HMGB1), one of the most important mediators of neuroinflammation, in the cortical microglia and cerebrospinal fluids. Moreover, acetylation-dependent nuclear export of HMGB1 was involved in up-regulation and secretion of HMGB1 after sleep deprivation. Further studies indicated that sleep deprivation induced an increase in the expression of acetyltransferase p300 and a decrease in the expression of deacetylase SIRT1, which synergistically enhanced the acetylation level of HMGB1 in the cortical microglial cells, thereby triggered the nuclear export and secretion of HMGB1. Most importantly, circadian clock protein PER2 constitutively interacted with p300 and inhibited its expression in the microglial cells, which can be interrupted by PER2 downregulation upon sleep deprivation, leading to the increased expression of p300 and acetylation and secretion of HMGB1. The truncated PER2 mutant without p300 binding ability lost its ability to regulate p300 expression, indicating that PER2 functioned as a co-suppressor of p300 in regulating acetylation and expression of HMGB1. Taken together, data in this study reveal a new mechanism by which PER2 is involved in controlling HMGB1 dependent neuroinflammation induced by sleep deprivation. Maintaining PER2 levels or blocking HMGB1 acetylation in the cortex might be prospective for preventing sleep deprivation-induced neuroinflammation and the related adverse reactions in the brain.
Collapse
Affiliation(s)
- Min Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhuoyao Ma
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Haoran Cui
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yumeng Miao
- Beijing Institute of Basic Medical Sciences, Beijing, China
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Yu Yin
- Beijing Institute of Basic Medical Sciences, Beijing, China
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qing Wen
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zhihui Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
- College of Life Science, Henan Normal University, Xinxiang, China
| | - Xin Huang
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Chen Xing
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Kun Liu
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Hui Peng
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lun Song
- Beijing Institute of Basic Medical Sciences, Beijing, China.
- Anhui Medical University, Hefei, China.
- College of Life Science, Henan Normal University, Xinxiang, China.
- School of Pharmacy, Jiamusi University, Jiamusi, China.
| |
Collapse
|
7
|
Horbatok K, Semchuk I, Horbach O, Khranovska N, Kosach V, Borysko P, Koniev S, Ulrich AS, Afonin S, Komarov IV. In vitro evaluation of the immunogenic potential of gramicidin S and its photocontrolled analogues. RSC Med Chem 2025:d5md00075k. [PMID: 40270993 PMCID: PMC12013366 DOI: 10.1039/d5md00075k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Three hallmarks of ICD (immunogenic cell death), release of adenosine triphosphate (ATP), release of high mobility group box 1 protein, and calreticulin exposure on the cell surface, were studied upon treatment of mammalian cells with small cyclic peptides, namely, the natural antibiotic gramicidin S (GS) and two photocontrolled GS analogues (LMB002 and LMB033). The analogues contained a photoisomerizable diarylethene fragment, and they exhibited different bioactivities in their "open" and "closed" photoisomeric forms. The data (obtained from cell cultures and spheroids) were collected in a concentration-dependent manner to assess cytotoxicity. Results showed that treatment with all peptides induced ICD at sub-IC50 and higher concentrations, indicating that GS and its derivatives have promising immunogenic potential. The "open" photoisomers of the photoswitchable GS analogues generated using visible light were as efficient as ICD inducers and the parent GS, while the UV-generated "closed" photoforms induced ICD only at higher concentrations. Herein, the cell specificity and time dependency of the observed effects are presented.
Collapse
Affiliation(s)
- Kateryna Horbatok
- Taras Shevchenko National University of Kyiv Volodymyrska street 60 01601 Kyiv Ukraine
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
| | - Iryna Semchuk
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | - Oleksandr Horbach
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | - Natalia Khranovska
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | | | - Petro Borysko
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
- V. P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry Akademician Kukhar street 1 02094 Kyiv Ukraine
| | - Serhii Koniev
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Anne S Ulrich
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Sergii Afonin
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Igor V Komarov
- Taras Shevchenko National University of Kyiv Volodymyrska street 60 01601 Kyiv Ukraine
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
- Lumobiotics Auerstraße 2 76227 Karlsruhe Germany
| |
Collapse
|
8
|
Shokr MM, Eladawy RM. HMGB1: Different secretion pathways with pivotal role in epilepsy and major depressive disorder. Neuroscience 2025; 570:55-67. [PMID: 39970982 DOI: 10.1016/j.neuroscience.2025.02.023] [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: 07/30/2024] [Revised: 11/10/2024] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
High-mobility group box 1 (HMGB1) protein is a highly prevalent protein that, once it is translocated to an extracellular site, can contribute to the pathogenesis of autoimmune and inflammatory responses, including epilepsy and depression. The conditions needed for release are associated with the production of multiple isoforms, and this translocation may occur in response to both immune cell activation and cell death. HMGB1 has been shown to interact with different mediators, including exportin 1, notch receptors, mitogen-activated protein kinase, STAT, tumor protein 53, and inflammasomes. Furthermore, as a crucial inflammatory mediator, HMGB1 has demonstrated upregulated expression and a higher percentage of translocation from the nucleus to the cytoplasm, acting on downstream receptors such as toll-like receptor 4 and receptor for advanced glycation end products, thereby activating interleukin-1 beta and nuclear factor kappa-B, intensifying inflammatory responses. In this review, we aim to discuss the different molecular interactions for the secretion of HMGB1 along with its pivotal role in epilepsy and major depressive disorder.
Collapse
Affiliation(s)
- Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University - Arish Branch, 45511 Arish, Egypt.
| | - Reem M Eladawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University - Arish Branch, 45511 Arish, Egypt
| |
Collapse
|
9
|
Biswas I, Panicker SR, Lupu F, Rezaie AR. Physiological significance of antithrombin D-helix interaction with vascular GAGs. Blood Adv 2025; 9:966-978. [PMID: 39671300 PMCID: PMC11907474 DOI: 10.1182/bloodadvances.2024014756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 12/15/2024] Open
Abstract
ABSTRACT Antithrombin (AT) is an anticoagulant serpin involved in the regulation of proteolytic activities of coagulation proteases. AT also possesses a direct anti-inflammatory function. The anticoagulant function of AT is mediated through its reactive center loop-dependent inhibition of coagulation proteases, but anti-inflammatory function of AT is mediated via its D-helix-dependent interaction with vascular glycosaminoglycans (GAGs). In vitro assays have established that therapeutic heparins promote the anticoagulant function of AT by binding D-helix and activating the serpin, however, the contribution of vascular GAGs to D-helix-dependent anticoagulant function of AT has remained poorly understood in vivo. Here, we explored this question by using 2 AT mutants, (AT-4Mut), which exhibits neither affinity for heparin nor D-helix-dependent anti-inflammatory signaling but possesses normal protease-inhibitory function and an inactive signaling-selective AT mutant in which its P1-Arg425 is deleted (AT-R425del). In vivo properties of mutants were compared with wild-type AT (AT-WT) in a small interfering RNA (siRNA)-mediated AT-deficient mouse model. The siRNA knockdown efficiently reduced expression of AT and induced robust procoagulant and proinflammatory phenotypes in mice. Infusion of both AT-WT and AT-4Mut rescued the procoagulant phenotype of AT-deficient mice as evidenced by restoration of the plasma clotting time and inhibition of fibrin deposition. AT-WT also attenuated inflammation as evidenced by reduced VCAM-1 expression and leukocyte infiltration in the liver and lungs; however, AT-4Mut failed to attenuate inflammation. Interestingly, AT-R425del also effectively attenuated inflammation in AT-depleted mice. These results suggest that interaction of AT D-helix with vascular GAGs may primarily be responsible for anti-inflammatory signaling rather than protease-inhibitory function of the serpin.
Collapse
Affiliation(s)
- Indranil Biswas
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Sumith R. Panicker
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Florea Lupu
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
| | - Alireza R. Rezaie
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK
- Department of Biochemistry and Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK
| |
Collapse
|
10
|
Gupta I, Patel AK. Deciphering HMGB1: Across a spectrum of DNA and nucleosome dynamics. Cell Biol Int 2025; 49:235-249. [PMID: 39551968 DOI: 10.1002/cbin.12260] [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: 06/14/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 11/19/2024]
Abstract
HMGB1 is the most abundant nonhistone nuclear protein, which has been widely studied for its roles in the cytoplasm as an autophagy mediator and in the extracellular matrix as an inflammatory molecule. Studies concerning HMGB1's actual role and its binding within the nucleus are inadequate. Through this in vitro study, we aimed to discern the binding parameters of HMGB1 with various types of DNA, nucleosomes, and chromatin. HMGB1 binds differentially to different DNA, with a high affinity for altered DNA structures such as triplex and bulge DNA. Remodelling of nucleosome by CHD7 remodeller was negatively impacted by the binding of HMGB1. We also found that HMGB1 binds to the linker DNA of chromatin. Findings from this study shed light on the diverse roles HMGB1 may play in transcription, gene expression, viral replication, CHARGE syndrome and so forth.
Collapse
Affiliation(s)
- Ishu Gupta
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Delhi, India
| | - Ashok K Patel
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, Delhi, India
| |
Collapse
|
11
|
Tanase DM, Valasciuc E, Gosav EM, Floria M, Buliga-Finis ON, Ouatu A, Cucu AI, Botoc T, Costea CF. Enhancing Retinal Resilience: The Neuroprotective Promise of BDNF in Diabetic Retinopathy. Life (Basel) 2025; 15:263. [PMID: 40003672 PMCID: PMC11856995 DOI: 10.3390/life15020263] [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: 01/09/2025] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Diabetic retinopathy (DR), a leading cause of vision impairment worldwide, is characterized by progressive damage to the retina due to prolonged hyperglycemia. Despite advances in treatment, current interventions largely target late-stage vascular complications, leaving underlying neurodegenerative processes insufficiently addressed. This article explores the crucial role in neuronal survival, axonal growth, and synaptic plasticity and the neuroprotective potential of Brain-Derived Neurotrophic Factor (BDNF) as a therapeutic strategy for enhancing retinal resilience in DR. Furthermore, it discusses innovative delivery methods for BDNF, such as gene therapy and nanocarriers, which may overcome the challenges of achieving sustained and targeted therapeutic levels in the retina, focusing on early intervention to preserve retinal function and prevent vision loss.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Oana Nicoleta Buliga-Finis
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.M.T.); (E.V.); (E.M.G.); (O.N.B.-F.); (A.O.)
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iasi, Romania
| | - Andrei Ionut Cucu
- Department of Biomedical Sciences, Faculty of Medicine and Biological Sciences, “Ștefan cel Mare” University, 720229 Suceava, Romania;
- Department of Neurosurgery, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Tina Botoc
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.B.); (C.F.C.)
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| | - Claudia Florida Costea
- Department of Ophthalmology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (T.B.); (C.F.C.)
- 2nd Ophthalmology Clinic, “Prof. Dr. Nicolae Oblu” Emergency Clinical Hospital, 700309 Iasi, Romania
| |
Collapse
|
12
|
Liu H, Liao X, Zhang Z, Min Q, Li Y, Xiong J, Lv Q, Xie X, Zhou J, Liao Z, Zhou H. HMGB1: key mediator in digestive system diseases. Inflamm Res 2025; 74:34. [PMID: 39903246 DOI: 10.1007/s00011-025-02002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025] Open
Abstract
High Mobility Group Box 1 (HMGB1), a multifunctional non-histone protein, and its involvement in various physiological and pathological contexts has garnered significant attention. Given HMGB1's central function in modulating key biological activities, such as inflammatory responses and cellular death, its contribution to the pathogenesis of digestive system diseases has become a focus of growing interest. This review aims to comprehensively explore the mechanisms by which HMGB1 contributes to the progression of inflammatory bowel disease (IBD), liver disorders, and pancreatitis. Furthermore, we explore the prospective clinical applications and outline future research directions for HMGB1 in digestive diseases, providing fresh perspectives that highlight the necessity of ongoing studies to understand its role in these conditions.
Collapse
Affiliation(s)
- Hengqian Liu
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, No. 181 Hanyu Road, Shapingba District, Chongqing, China
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Xiping Liao
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Zuo Zhang
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Qian Min
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Yuanyuan Li
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Junzhi Xiong
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Qiao Lv
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Xia Xie
- Department of Gastroenterology, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| | - Jianyun Zhou
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China
| | - Zhongli Liao
- School of Medicine, Chongqing University Cancer Hospital, Chongqing University, No. 181 Hanyu Road, Shapingba District, Chongqing, China.
| | - Hongli Zhou
- Clinical Medical Research Center, The Second Affiliated Hospital, Army Medical University, No. 83 Xinqiao Main Street, Shapingba District, Chongqing, China.
| |
Collapse
|
13
|
Chen Z, Ruan F, Wu D, Yu X, Jiang Y, Bao W, Wen H, Hu J, Bi H, Chen L, Le K. Quercetin alleviates neonatal hypoxic-ischaemic brain injury by rebalancing microglial M1/M2 polarization through silent information regulator 1/ high mobility group box-1 signalling. Inflammopharmacology 2025; 33:865-883. [PMID: 39565473 DOI: 10.1007/s10787-024-01599-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/02/2024] [Indexed: 11/21/2024]
Abstract
Neonatal hypoxic-ischaemic encephalopathy (HIE) remains one of the major causes of neonatal death and long-term neurological disability. Due to its complex pathogenesis, there are still many challenges in its treatment. In our previous studies, we found that quercetin can alleviate neurological dysfunction after hypoxic-ischaemic brain injury (HIBI) in neonatal mice. As demonstrated through in vitro experiments, quercetin can inhibit the activation of the TLR4/MyD88/NF-κB signalling pathway and the inflammatory response in the microglial cell line BV2 after oxygen-glucose deprivation. However, the in-depth mechanism still needs to be further elucidated. In the present study, 7 day-old neonatal ICR mice or BV2 cells were treated with quercetin with or without the SIRT1 inhibitor EX527 via neurobehavioural, histopathological and molecular methods. In vivo experiments have shown that quercetin can significantly improve the performance of HI mice in behavioural tests, such as the Morris water maze, rotarod test and pole climbing test, and reduce HI insult-induced structural brain damage, cell apoptosis and hippocampal neuron loss. Quercetin also inhibited the immunofluorescence intensity of the microglial M1 marker CD16 + 32 and significantly downregulated the expression of the M1-related proteins iNOS, IL-1β and TNF-α. Moreover, quercetin increased the immunofluorescence intensity of the microglial M2 marker CD206 and significantly increased the expression of the M2-related proteins Arg-1 and IL-10. In addition, quercetin limits the nucleocytoplasmic translocation and release of microglial HMGB1 and further suppresses the activation of the downstream TLR4/MyD88/NF-κB signalling pathway. The above effects of quercetin are partially weakened by pretreatment with EX527. Similar results were found in in vitro experiments, and the mechanism further revealed that the rebalancing effect of quercetin on microglial polarization is achieved through the SIRT1-mediated reduction in HMGB1 acetylation levels. This study provides new and complementary insights into the neuroprotective effects of quercetin and a new direction for the treatment of neonatal HIE.
Collapse
Affiliation(s)
- Zhaoyan Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Fei Ruan
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Di Wu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Xiaoping Yu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Yaqing Jiang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Wei Bao
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Haicheng Wen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Jing Hu
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Haidi Bi
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China
| | - Liping Chen
- Department of Neonatology, Jiangxi Children's Hospital, No.122 Yangming Road, Nanchang, 330006, Jiangxi Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.17 Yongwaizheng Street, Nanchang, 330006, Jiangxi Province, China.
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, 11 Yuk Choi Rd, Hong Kong S.A.R., Hong Kong, China.
| |
Collapse
|
14
|
Date S, Bhatt LK. Targeting high-mobility-group-box-1-mediated inflammation: a promising therapeutic approach for myocardial infarction. Inflammopharmacology 2025; 33:767-784. [PMID: 39487941 DOI: 10.1007/s10787-024-01586-w] [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: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Myocardial ischemia, resulting from coronary artery blockage, precipitates cardiac arrhythmias, myocardial structural changes, and heart failure. The pathophysiology of MI is mainly based on inflammation and cell death, which are essential in aggravating myocardial ischemia and reperfusion injury. Emerging research highlights the functionality of high mobility group box-1, a non-histone nucleoprotein functioning as a chromosomal stabilizer and inflammatory mediator. HMGB1's release into the extracellular compartment during ischemia acts as damage-associated molecular pattern, triggering immune reaction by pattern recognition receptors and exacerbating tissue inflammation. Its involvement in signaling pathways like PI3K/Akt, TLR4/NF-κB, and RAGE/HMGB1 underscores its significance in promoting angiogenesis, apoptosis, and reducing inflammation, which is crucial for MI treatment strategies. This review highlights the complex function of HMGB1 in the pathogenesis of myocardial infarction by summarizing novel findings on the protein in ischemic situations. Understanding the mechanisms underlying HMGB1 could widen the way to specific treatments that minimize the severity of MI and enhance patient outcomes.
Collapse
Affiliation(s)
- Shrutika Date
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
15
|
Sen MG, Chooi R, McMullen JR. Heart-derived factors and organ cross-talk in settings of health and disease: new knowledge and clinical opportunities for multimorbidity. J Physiol 2025. [PMID: 39888058 DOI: 10.1113/jp287400] [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: 09/26/2024] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Cardiovascular disease affects millions of people worldwide and often presents with other conditions including metabolic, renal and neurological disorders. A variety of secreted factors from multiple organs/tissues (proteins, nucleic acids and lipids) have been implicated in facilitating organ cross-talk that may contribute to the development of multimorbidity. Secreted proteins have received the most attention, with the greatest body of research related to factors released from adipose tissue (adipokines), followed by skeletal muscle (myokines). To date, there have been fewer studies on proteins released from the heart (cardiokines) implicated with organ cross-talk. Early evidence for the secretion of cardiac-specific factors facilitating organ cross-talk came in the form of natriuretic peptides which are secreted via the classical endoplasmic reticulum-Golgi pathway. More recently, studies in cardiomyocyte-specific genetic mouse models have revealed cardiac-initiated organ cross-talk. Cardiomyocyte-specific modulation of microRNAs (miR-208a and miR-23-27-24 cluster) and proteins such as the mediator complex subunit 13 (MED13), G-protein-coupled receptor kinase 2 (GRK2), mutant α-myosin heavy-chain (αMHC), ubiquitin-like modifier-activating enzyme (ATG7), oestrogen receptor alpha (ERα) and fibroblast growth factor 21 (FGF21) have resulted in metabolic and renal phenotypes. These studies have implicated a variety of factors which can be secreted via the classical pathway or via non-classical mechanisms including the release of extracellular vesicles. Cross-talk between the heart and the brain has also been described (e.g. via miR-1 and an emerging concept, interoception: detection of internal neural signals). Here we summarize these studies taking into consideration that factors may be secreted in both settings of health and in disease.
Collapse
Affiliation(s)
- Melodi G Sen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Roger Chooi
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Heart Research Institute, Newtown, New South Wales, Australia
- Monash Alfred Baker Centre for Cardiovascular Research, Faculty of Medicine Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
- Baker Department of Cardiometabolic Health, The University of Melbourne, Parkville, Victoria, Australia
- Baker Department of Cardiovascular Research, Translation and Implementation, La Trobe University, Bundoora, Victoria, Australia
| |
Collapse
|
16
|
Cheng J, Tian X, Wu C, Wang J, Liu H, Cheng S, Sun H. MiR- 146b-5p inhibits Candida albicans-induced inflammatory response through targeting HMGB1 in mouse primary peritoneal macrophages. Heliyon 2025; 11:e41464. [PMID: 39844980 PMCID: PMC11751530 DOI: 10.1016/j.heliyon.2024.e41464] [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: 05/05/2024] [Revised: 08/21/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Background Candida albicans (C. albicans) is one of the most common pathogens associated with deep fungal infection, which represents a serious threat to human health. Although high mobility group box 1 (HMGB1) plays a key role in C. albicans infection, its mechanism is unclear. We aimed to explore the regulation of small-molecule non-coding RNA (miRNA) for HMGB1 in C. albicans infection. Methods Mouse primary peritoneal macrophages (MPMs) were isolated successfully. The optimum conditions for C. albicans infection were selected by Western blot and ELISA. The miRNA differential expression profiles of C. albicans infection were screened and verified by 6 miRNA gene chips and qRT-PCR. The direct regulation of the target gene HMGB1 by mmu-miR-146b-5p was confirmed through a dual-luciferase assay. The levels of mmu-miR-146b-5p, HMGB1, inflammatory mediators, p-IKK, IKK, p-IκBα, IκBα and NF-κB p65 were tested by qRT-PCR, Western blot, and ELISA. The nuclear and cytoplasm translocation of HMGB1 and NF-κB p65 were detected by Western blot and laser confocal microscopy. After siHMGB1 transfection, the expression levels of HMGB1, inflammatory mediators, p-IKK, IKK, p-IκBα, IκBα and NF-κB p65 were assessed using Western blot, qRT-PCR and ELISA. Results In our study, MPMs were successfully extracted and infected with C. albicans at optimum conditions of 1.5 × 107 CFU/mL for 36 h. Through miRNA gene chips analysis, 40 differential genes were screened. mmu-miR-146b-5p could directly and negatively regulate the expression and translocation of HMGB1, inhibit the expression of inflammatory mediators, and might participate in the NF-κB signaling pathway in a HMGB1-dependent manner under C. albicans infection. Conclusion mmu-miR-146b-5p may play an anti-inflammatory role in treating C. albicans infection and provide a novel target for it.
Collapse
Affiliation(s)
- Jing Cheng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Health Care, Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Xiaoxing Tian
- The Fifth People's Hospital of Chongqing, Chongqing, China
| | - Chuanxin Wu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jiaojiao Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Huiling Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Sha Cheng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hang Sun
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
17
|
Saha A, Islam MM, Kumar R, Ismail AM, Garcia E, Gullapali RR, Chodosh J, Rajaiya J. Virus and Cell Specific HMGB1 Secretion and Subepithelial Infiltrate Formation in Adenovirus Keratitis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631509. [PMID: 39829903 PMCID: PMC11741304 DOI: 10.1101/2025.01.07.631509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
A highly contagious infection caused by human adenovirus species D (HAdV-D), epidemic keratoconjunctivitis (EKC) results in corneal subepithelial infiltration (SEI) by leukocytes, the hallmark of the infection. To date, the pathogenesis of corneal SEI formation in EKC is unresolved. HMGB1 (high-mobility group box 1 protein) is an alarmin expressed in response to infection and a marker of sepsis. Earlier studies using a different adenovirus species, HAdV-C, showed retention of HMGB1 in the infected cell nucleus by adenovirus protein VII, enabling immune evasion. Here, using HAdV-D we show cell-specific HMGB1 secretion by infected cells, and provide an HAdV-D specific mechanism for SEI formation in EKC. HMGB1 was secreted only upon infection of human corneal epithelial cells, not from other cell types, and only upon infection by HAdV-D types associated with EKC. Acetylated HMGB1 translocation from the nucleus to the cytoplasm, then to the extracellular milieu, was tightly controlled by CRM1 and LAMP1, respectively. Primary stromal cells when stimulated by rHMGB1 expressed proinflammatory chemokines. In a novel 3D culture system in tune with the architecture of the cornea, HMGB1 released by infected corneal epithelial cells induced leukocytic infiltrates either directly and/or indirectly via stimulated stromal cells, which together explains SEI formation in EKC.
Collapse
|
18
|
Hollis A, Lukens JR. Role of inflammasomes and neuroinflammation in epilepsy. Immunol Rev 2025; 329:e13421. [PMID: 39523682 PMCID: PMC11744240 DOI: 10.1111/imr.13421] [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] [Indexed: 11/16/2024]
Abstract
Epilepsy is a brain disorder characterized by recurrent seizures, which are brief episodes of abnormal electrical activity in the brain and involuntary movement that can lead to physical injury and loss of consciousness. Seizures are canonically accompanied by increased inflammatory cytokine production that promotes neuroinflammation, brain pathology, and seizure propagation. Understanding the source of pro-inflammatory cytokines which promote seizure pathogenesis could be a gateway to precision epilepsy drug design. This review discusses the inflammasome in epilepsy including its role in seizure propagation and negative impacts on brain health. The inflammasome is a multiprotein complex that coordinates IL-1β and IL-18 production in response to tissue damage, cellular stress, and infection. Clinical evidence for inflammasome signaling in epileptogenesis is reviewed followed by a discussion of emerging strategies to modulate inflammasome activity in epilepsy.
Collapse
Affiliation(s)
- Ava Hollis
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of NeuroscienceUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
19
|
Wang L, Dong Z, Zhang Y, Peng L. Emerging Roles of High-mobility Group Box-1 in Liver Disease. J Clin Transl Hepatol 2024; 12:1043-1056. [PMID: 39649031 PMCID: PMC11622203 DOI: 10.14218/jcth.2024.00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 12/10/2024] Open
Abstract
High-mobility group box-1 (HMGB1) is an architectural chromosomal protein with various roles depending on its cellular localization. Extracellular HMGB1 functions as a prototypical damage-associated molecular pattern that triggers inflammation and adaptive immune responses, mediated by specific cell surface receptors, including receptors for advanced glycation end products and toll-like receptors. Post-translational modifications of HMGB1 significantly impact various cellular processes that contribute to the pathogenesis of liver diseases. Recent studies have highlighted the close relationship between HMGB1 and the pathogenesis of acute liver injuries, including acetaminophen-induced liver injury, hepatic ischemia-reperfusion injury, and acute liver failure. In chronic liver diseases, HMGB1 plays a role in nonalcoholic fatty liver disease, alcohol-associated liver disease, liver fibrosis, and hepatocellular carcinoma. Targeting HMGB1 as a therapeutic approach, either by inhibiting its release or blocking its extracellular function, is a promising strategy for treating liver diseases. This review aimed to summarize the available evidence on HMGB1's role in liver disease, focusing on its multifaceted signaling pathways, impact on disease progression, and the translation of these findings into clinical interventions.
Collapse
Affiliation(s)
- Lu Wang
- Department of Diagnostics, Second School of Clinical Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zhiwei Dong
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yeqiong Zhang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
20
|
Shen P, Zhang L, Jiang X, Yu B, Zhang J. Targeting HMGB1 and Its Interaction with Receptors: Challenges and Future Directions. J Med Chem 2024; 67:21671-21694. [PMID: 39648929 DOI: 10.1021/acs.jmedchem.4c01912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
High mobility group box 1 (HMGB1) is a nonhistone chromatin protein predominantly located in the nucleus. However, under pathological conditions, HMGB1 can translocate from the nucleus to the cytoplasm and subsequently be released into the extracellular space through both active secretion and passive release mechanisms. The distinct cellular locations of HMGB1 facilitate its interaction with various endogenous and exogenous factors, allowing it to perform diverse functions across a range of diseases. This Perspective provides a comprehensive overview of the structure, release mechanisms, and multifaceted roles of HMGB1 in disease contexts. Furthermore, it introduces the development of both small molecule and macromolecule inhibitors targeting HMGB1 and its interaction with receptors. A detailed analysis of the predicted pockets is also presented, aiming to establish a foundation for the future design and development of HMGB1 inhibitors.
Collapse
Affiliation(s)
- Pingping Shen
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Libang Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xuewa Jiang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| | - Jian Zhang
- Department of Resources Science of Traditional Chinese Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, P. R. China
| |
Collapse
|
21
|
Li J, Lou L, Chen W, Qiang X, Zhu C, Wang H. Connexin 43 and Pannexin 1 hemichannels as endogenous regulators of innate immunity in sepsis. Front Immunol 2024; 15:1523306. [PMID: 39763679 PMCID: PMC11701031 DOI: 10.3389/fimmu.2024.1523306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/05/2024] [Indexed: 02/02/2025] Open
Abstract
Sepsis is a life-threatening organ dysfunction resulting from a dysregulated host response to infections that is initiated by the body's innate immune system. Nearly a decade ago, we discovered that bacterial lipopolysaccharide (LPS) and serum amyloid A (SAA) upregulated Connexin 43 (Cx43) and Pannexin 1 (Panx1) hemichannels in macrophages. When overexpressed, these hemichannels contribute to sepsis pathogenesis by promoting ATP efflux, which intensifies the double-stranded RNA-activated protein kinase R (PKR)-dependent inflammasome activation, pyroptosis, and the release of pathogenic damage-associated molecular pattern (DAMP) molecules, such as HMGB1. Mimetic peptides targeting specific regions of Cx43 and Panx1 can distinctly modulate hemichannel activity in vitro, and diversely impact sepsis-induced lethality in vivo. Along with extensive supporting evidence from others, we now propose that hemichannel molecules play critical roles as endogenous regulators of innate immunity in sepsis.
Collapse
Affiliation(s)
- Jianhua Li
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Li Lou
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Weiqiang Chen
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Xiaoling Qiang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Cassie Zhu
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| | - Haichao Wang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Emergency Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, United States
| |
Collapse
|
22
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMID: 39766257 PMCID: PMC11673996 DOI: 10.3390/biom14121550] [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: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
23
|
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
|
24
|
Hisaoka-Nakashima K, Takeuchi Y, Saito Y, Shimoda T, Nakamura Y, Wang D, Liu K, Nishibori M, Morioka N. Glucocorticoids induce HMGB1 release in primary cultured rat cortical microglia. Neuroscience 2024; 560:56-66. [PMID: 39304023 DOI: 10.1016/j.neuroscience.2024.09.031] [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: 06/30/2024] [Revised: 09/01/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
Stress, a risk factor for major depressive disorder and Alzheimer disease, leads to the release of high-mobility group box-1 (HMGB1) protein, which in turn causes neuroinflammation. The mechanism underlying stress-induced HMGB1 release is unknown, but stress-associated glucocorticoids could be involved. Primary cultured rat cortical microglia and neurons were treated with corticosterone, a stress-associated glucocorticoid, and HMGB1 release was measured by ELISA and western blotting to test this hypothesis. With corticosterone treatment, significant HMGB1 was released in microglia but not in neuronal cell cultures. HMGB1 mRNA expression and HMGB1 protein expression in microglia were not affected by corticosterone treatment. Thus, the source of extracellular HMGB1 released into the medium is likely to be existing nuclear HMGB1 rather than newly synthesized HMGB1. Corticosterone-induced HMGB1 release in microglia culture was significantly attenuated by blocking glucocorticoid receptors but not mineralocorticoid receptors. Dexamethasone, a selective glucocorticoid receptor agonist, and dexamethasone-bovine serum albumin (BSA), a membrane-impermeable glucocorticoid receptor agonist used to confirm the membrane receptor-mediated effects of glucocorticoids, increased the release of HMGB1. Immunocytochemistry showed that HMGB1 translocated from the nucleus to the cytoplasm following dexamethasone or dexamethasone-BSA treatment through glucocorticoid receptors. The present findings suggest that glucocorticoids stimulate microglial membrane glucocorticoid receptors and trigger cytoplasmic translocation and extracellular release of nuclear HMGB1. Thus, under stress conditions, glucocorticoids induce microglial HMGB1 release, leading to a neuroinflammatory state that could mediate neurological disorders.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yuka Takeuchi
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yukino Saito
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Takahisa Shimoda
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan; Laboratory for Pharmacology, Pharmaceuticals Research Center, Asahi Kasei Pharma Co. Ltd., 632-1 Mifuku, Izunokuni, Shizuoka 410-2321, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Dengli Wang
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Keyue Liu
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Masahiro Nishibori
- Department of Translational Research & Drug Development, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
25
|
Xing WQ, Piao XJ, Han Q, Shi HY, Wu WC, Si F, Lu JJ, Zhou TZ, Guo JR, Li SZ, Xu B. SIRT2 regulates high mobility group protein B1 nucleoplasmic shuttle and degradation via deacetylation in microglia. J Cell Physiol 2024; 239:e31364. [PMID: 39129208 DOI: 10.1002/jcp.31364] [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: 03/02/2024] [Revised: 06/13/2024] [Accepted: 06/19/2024] [Indexed: 08/13/2024]
Abstract
High mobility group protein B1 (HMGB1) acts as a pathogenic inflammatory response to mediate ranges of conditions such as epilepsy, septic shock, ischemia, traumatic brain injury, Parkinson's disease, Alzheimer's disease and mass spectrometry. HMGB1 promotes inflammation during sterile and infectious damage and plays a crucial role in disease development. Mobilization from the nucleus to the cytoplasm is the first important step in the release of HMGB1 from activated immune cells. Here, we demonstrated that Sirtuin 2 (SIRT2) physically interacts with and deacetylates HMGB1 at 43 lysine residue at nuclear localization signal locations, strengthening its interaction with HMGB1 and causing HMGB1 to be localized in the cytoplasm. These discoveries are the first to shed light on the SIRT2 nucleoplasmic shuttle, which influences HMGB1 and its degradation, hence revealing novel therapeutic targets and avenues for neuroinflammation treatment.
Collapse
Affiliation(s)
- Wan-Qun Xing
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Xian-Ji Piao
- The Fifth Affiliated Hospital of Harbin Medical University, Daqing, China
| | - Qi Han
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Hui-Ying Shi
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Wen-Cong Wu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Fan Si
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Jing-Jing Lu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Tie-Zhong Zhou
- College of Animal Husbandry and Veterinary Medicine, Jinzhou Medical University, Jinzhou, China
| | - Jing-Ru Guo
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shi-Ze Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Bin Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| |
Collapse
|
26
|
Ge X, Subramaniyam N, Song Z, Desert R, Han H, Das S, Komakula SSB, Wang C, Lantvit D, Ge Z, Hoshida Y, Nieto N. Post-translational modifications drive the effects of HMGB1 in alcohol-associated liver disease. Hepatol Commun 2024; 8:e0549. [PMID: 39760999 PMCID: PMC11495752 DOI: 10.1097/hc9.0000000000000549] [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: 07/12/2024] [Accepted: 08/26/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND We previously identified that high-mobility group box-1 (HMGB1) is increased and undergoes post-translational modifications (PTMs) in response to alcohol consumption. Here, we hypothesized that specific PTMs, occurring mostly in hepatocytes and myeloid cells, could contribute to the pathogenesis of alcohol-associated liver disease (AALD). METHODS We used the Lieber-DeCarli (LD) model of early alcohol-induced liver injury, combined with engineered viral vectors and genetic approaches to regulate the expression of HMGB1, its PTMs (reduced [H], oxidized [O], acetylated [Ac], both [O + Ac]), and its receptors (RAGE, TLR4) in a cell-specific manner (hepatocytes and/or myeloid cells). RESULTS Hmgb1 ablation in hepatocytes or myeloid cells partially protected, while ablation in both prevented steatosis, inflammation, IL1B production, and alcohol-induced liver injury. Hepatocytes were a major source of [H], [O], and [Ac] HMGB1, whereas myeloid cells produced only [H] and [Ac] HMGB1. Neutralization of HMGB1 prevented, whereas injection of [H] HMGB1 increased AALD, which was worsened by injection of [O] HMGB1. While [O] HMGB1 induced liver injury, [Ac] HMGB1 protected and counteracted the effects of [O] HMGB1 in AALD. [O] HMGB1 stimulated macrophage (MF) migration, activation, IL1B production, and secretion. Ethanol-fed RageΔMye but not Tlr4ΔMye, RageΔHep, or Tlr4ΔHep mice were protected from AALD, indicating a crucial role of RAGE in myeloid cells for AALD. [O] HMGB1 recruited and activated myeloid cells through RAGE and contributed to steatosis, inflammation, and IL1B production in AALD. CONCLUSIONS These results provide evidence for targeting [O] HMGB1 of hepatocyte origin as a ligand for RAGE signaling in myeloid cells and a driver of steatosis, inflammatory cell infiltration, and IL1B production in AALD. Importantly, we reveal that [Ac] HMGB1 offsets the noxious effects of [O] HMGB1 in AALD.
Collapse
Affiliation(s)
- Xiaodong Ge
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | | | - Zhuolun Song
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Romain Desert
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Hui Han
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Sukanta Das
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | | | - Chao Wang
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Daniel Lantvit
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Zhiyan Ge
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Yujin Hoshida
- Department of Internal Medicine, Division of Digestive and Liver Diseases, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Natalia Nieto
- Department of Pathology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois Chicago, Chicago, Illinois, USA
- Research & Development Service, Jesse Brown Veterans Affairs Medical Center, Chicago, lllinois, USA
| |
Collapse
|
27
|
Wasim R, Singh A, Islam A, Mohammed S, Anwar A, Mahmood T. High Mobility Group Box 1 and Cardiovascular Diseases: Study of Act and Connect. Cardiovasc Toxicol 2024; 24:1268-1286. [PMID: 39242448 DOI: 10.1007/s12012-024-09919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Cardiovascular disease is the deadly disease that can result in sudden death, and inflammation plays an important role in its onset and progression. High mobility group box 1 (HMGB1) is a nuclear protein that regulates transcription, DNA replication, repair, and nucleosome assembly. HMGB1 is released passively by necrotic tissues and actively secreted by stressed cells. Extracellular HMGB1 functions as a damage associated molecular patterns molecule, producing numerous redox forms that induce a range of cellular responses by binding to distinct receptors and interactors, including tissue inflammation and regeneration. Extracellular HMGB1 inhibition reduces inflammation and is protective in experimental models of myocardial ischemia/reperfusion damage, myocarditis, cardiomyopathies caused by mechanical stress, diabetes, bacterial infection, or chemotherapeutic drugs. HMGB1 administration following a myocardial infarction followed by permanent coronary artery ligation improves cardiac function by stimulating tissue regeneration. HMGB1 inhibits contractility and produces hypertrophy and death in cardiomyocytes, while also stimulating cardiac fibroblast activity and promoting cardiac stem cell proliferation and differentiation. Maintaining normal nuclear HMGB1 levels, interestingly, protects cardiomyocytes from apoptosis by limiting DNA oxidative stress, and mice with HMGB1cardiomyocyte-specific overexpression are partially protected from cardiac injury. Finally, elevated levels of circulating HMGB1 have been linked to human heart disease. As a result, following cardiac damage, HMGB1 elicits both detrimental and helpful responses, which may be due to the formation and stability of the various redox forms, the particular activities of which in this context are mostly unknown. This review covers recent findings in HMGB1 biology and cardiac dysfunction.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacy, Integral University, Lucknow, 226026, India.
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Aditya Singh
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Anas Islam
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Saad Mohammed
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Aamir Anwar
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| | - Tarique Mahmood
- Department of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
28
|
Vladimirova D, Staneva S, Ugrinova I. Multifaceted role of HMGB1: From nuclear functions to cytoplasmic and extracellular signaling in inflammation and cancer-Review. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 143:271-300. [PMID: 39843137 DOI: 10.1016/bs.apcsb.2024.09.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
High-mobility group box 1 (HMGB1) is a highly conserved nuclear protein involved in key nuclear processes such as DNA repair, replication, and gene regulation. Beyond its established nuclear roles, HMGB1 has crucial functions in the cytoplasm and extracellular environment. When translocated to the cytoplasm, HMGB1 plays a role in autophagy, cell survival, and immune response modulation. In its extracellular form, HMGB1 acts as a damage-associated molecular pattern molecule, initiating inflammatory responses by interacting with receptors such as Receptor for advanced glycation endproducts and Toll-like receptors. Recent studies have shown its role in promoting tissue regeneration, wound healing, and angiogenesis, highlighting its dual role in both inflammation and tissue repair. Notably, the redox status of HMGB1 influences its function, with the reduced form promoting autophagy and the disulfide form driving inflammation. Dysregulation of HMGB1 contributes to the progression of various diseases, including cancer, where it influences tumor growth, metastasis, and resistance to therapy. This review provides an overview of the nuclear, cytoplasmic, and extracellular roles of HMGB1, discussing its involvement in nuclear homeostasis, rare genetic diseases, autophagy, inflammation, cancer progression, and tissue regeneration.
Collapse
Affiliation(s)
- Desislava Vladimirova
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Sonya Staneva
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Iva Ugrinova
- "Roumen Tsanev" Institute of Molecular Biology, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| |
Collapse
|
29
|
Zeng M, Liang G, Yuan F, Yan S, Liu J, He Z. Macrophages-derived high-mobility group box-1 protein induces endothelial progenitor cells pyroptosis. iScience 2024; 27:110996. [PMID: 39421592 PMCID: PMC11483297 DOI: 10.1016/j.isci.2024.110996] [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: 01/09/2024] [Revised: 05/08/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024] Open
Abstract
Endothelial dysfunction is an important factor in the progress of sepsis. Endothelial progenitor cells (EPCs) are the precursor cells of endothelial cells and play a crucial role in the prognosis and treatment of sepsis. EPCs in the peripheral blood of patients with sepsis undergo pyroptosis, but the mechanism remains much of unknown. Serum high-mobility group box-1 (HMGB1) is significantly elevated in patients with sepsis, but whether it is related to EPCs pyroptosis is unknown. We used a cell model of sepsis in vitro to isolate EPCs for better observation. By detecting the pyroptosis-related indicators of EPCs and the level of release and acetylation of HMGB1 in inflammatory macrophages, it was found that HMGB1 released by inflammatory macrophages combined with receptor for advanced glycation end products (RAGE) is a key pathway to induce pyroptosis of EPCs.
Collapse
Affiliation(s)
- Menghao Zeng
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Changsha, Hunan, China
| | - Guibin Liang
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Changsha, Hunan, China
| | - Fangfang Yuan
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Shanshan Yan
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Jie Liu
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
| | - Zhihui He
- Department of Critical Care Medicine, the Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, China
- Sepsis Translational Medicine Key Laboratory of Hunan Province, Changsha, Hunan, China
- National Engineering Research Center for Human Stem Cells, Changsha, Hunan, China
| |
Collapse
|
30
|
Wu T, Lu Y, Yu Y, Hua Y, Ge G, Zhao W, Chen K, Zhong Z, Zhang F. Long noncoding RNA AK144717 exacerbates pathological cardiac hypertrophy through modulating the cellular distribution of HMGB1 and subsequent DNA damage response. Cell Mol Life Sci 2024; 81:432. [PMID: 39395058 PMCID: PMC11470913 DOI: 10.1007/s00018-024-05464-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/09/2024] [Accepted: 09/27/2024] [Indexed: 10/14/2024]
Abstract
DNA damage induced by oxidative stress during cardiac hypertrophy activates the ataxia telangiectasia mutated (ATM)-mediated DNA damage response (DDR) signaling, in turn aggravating the pathological cardiomyocyte growth. This study aims to identify the functional associations of long noncoding RNA (lncRNAs) with cardiac hypertrophy and DDR. The altered ventricular lncRNAs in the mice between sham and transverse aortic constriction (TAC) group were identified by microarray analysis, and a novel lncRNA AK144717 was found to gradually upregulate during the development of pathological cardiac hypertrophy induced by TAC surgery or angiotensin II (Ang II) stimulation. Silencing AK144717 had a similar anti-hypertrophic effect to that of ATM inhibitor KU55933 and also suppressed the activated ATM-DDR signaling induced by hypertrophic stimuli. The involvement of AK144717 in DDR and cardiac hypertrophy was closely related to its interaction with HMGB1, as silencing HMGB1 abolished the effects of AK144717 knockdown. The binding of AK144717 to HMGB1 prevented the interaction between HMGB1 and SIRT1, contributing to the increased acetylation and then cytosolic translocation of HMGB1. Overall, our study highlights the role of AK144717 in the hypertrophic response by interacting with HMGB1 and regulating DDR, hinting that AK144717 is a promising therapeutic target for pathological cardiac growth.
Collapse
Affiliation(s)
- Tianyu Wu
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Yao Lu
- Department of Cardiology, Xuzhou Central Hospital, The Xuzhou School of Clinical Medicine of Nanjing Medical University, No.199 Jiefang South Road, Xuzhou, 221009, PR China
| | - Yue Yu
- Department of Cardiology, Nanjing University Medical School Affiliated Nanjing Drum Tower Hospital, Zhongshan Road 321, Nanjing, 210029, PR China
| | - Yan Hua
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Gaoyuan Ge
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Wei Zhao
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Kaiyan Chen
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Zhuen Zhong
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China
| | - Fengxiang Zhang
- Section of Pacing and Electrophysiology, Division of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, PR China.
| |
Collapse
|
31
|
Chen L, Huang L, Gu Y, Li C, Sun P, Xiang Y. Novel post-translational modifications of protein by metabolites with immune responses and immune-related molecules in cancer immunotherapy. Int J Biol Macromol 2024; 277:133883. [PMID: 39033895 DOI: 10.1016/j.ijbiomac.2024.133883] [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: 07/01/2023] [Revised: 06/30/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Tumour immunotherapy is an effective and essential treatment for cancer. However, the heterogeneity of tumours and the complex and changeable tumour immune microenvironment (TME) creates many uncertainties in the clinical application of immunotherapy, such as different responses to tumour immunotherapy and significant differences in individual efficacy. It makes anti-tumour immunotherapy face many challenges. Immunometabolism is a critical determinant of immune cell response to specific immune effector molecules, significantly affecting the effects of tumour immunotherapy. It is attributed mainly to the fact that metabolites can regulate the function of immune cells and immune-related molecules through the protein post-translational modifications (PTMs) pathway. This study systematically summarizes a variety of novel protein PTMs including acetylation, propionylation, butyrylation, succinylation, crotonylation, malonylation, glutarylation, 2-hydroxyisobutyrylation, β-hydroxybutyrylation, benzoylation, lactylation and isonicotinylation in the field of tumour immune regulation and immunotherapy. In particular, we elaborate on how different PTMs in the TME can affect the function of immune cells and lead to immune evasion in cancer. Lastly, we highlight the potential treatment with the combined application of target-inhibited protein modification and immune checkpoint inhibitors (ICIs) for improved immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China
| | - Yu Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Chen Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China.
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China.
| |
Collapse
|
32
|
Bianchi ME, Rubartelli A, Sitia R. Preferential Secretion of Oxidation-Sensitive Proteins by Unconventional Pathways: Why is This Important for Inflammation? Antioxid Redox Signal 2024; 41:693-705. [PMID: 38916186 DOI: 10.1089/ars.2024.0554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Significance: Fidelity of intercellular communication depends on unambiguous interactions between protein ligands and membrane receptors. Most proteins destined to the extracellular space adopt the required three-dimensional shape as they travel through the endoplasmic reticulum (ER), Golgi complex, and other organelles of the exocytic pathway. However, some proteins, many of which are involved in inflammation, avoid this classical secretory route and follow unconventional pathways to leave the cell. Recent Advances: Stringent quality control systems operate in the ER and cis-Golgi, restricting transport to native conformers, devoid of non-native disulfides and/or reactive thiols. However, some proteins released by living cells require reduced cysteines to exert their extracellular function(s). Remarkably, these proteins lack the secretory signal sequence normally required by secretory proteins for translocation into the ER lumen. Critical Issues: Why do interleukin-1β, high mobility group box 1, and other proinflammatory proteins avoid the ER-Golgi route to reach the intercellular space? These proteins require reactive cysteines for exerting their function. Therefore, eluding thiol-mediated quality control along the exocytic pathway is likely one of the main reasons why extracellular proteins that need to be reduced utilize unconventional pathways of secretion, where a quality control aimed at oxidating native cysteines is not present. Future Directions: Particularly under stress conditions, cells release redox-active enzymes and nonprotein thiol compounds that exert an extracellular control of redox-sensitive protein activity, shaping inflammatory responses. This post-secretion, redox-dependent editing of protein messages is still largely undefined. Understanding the underlying mechanistic events will hopefully provide new tools to control inflammation. Antioxid. Redox Signal. 41, 693-705.
Collapse
Affiliation(s)
- Marco E Bianchi
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milano, Italy
| | - Anna Rubartelli
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milano, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Vita-Salute San Raffaele University and IRCCS San Raffaele Hospital, Milano, Italy
| |
Collapse
|
33
|
Mu S, Li Z, Lin L, Wang D, Yang F, Chen L, Xian L, Lin K, Lin Y, Ye D, Yang Y, Wei L, Xu Y, Wang S. SIRT1-Mediated HMGB1 Deacetylation Suppresses Neutrophil Extracellular Traps Related to Blood-Brain Barrier Impairment After Cerebral Venous Thrombosis. Mol Neurobiol 2024; 61:6060-6076. [PMID: 38267754 DOI: 10.1007/s12035-024-03959-2] [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: 07/26/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Cerebral venous thrombosis (CVT) is a neurovascular disease with recently increasing incidence. Aseptic inflammatory responses play an important role in the pathology of CVT. Recent studies report that neutrophil extracellular traps (NETs) are major triggers of thrombosis and inflammation in stroke, but their effect on brain injury in CVT requires further validation. In this study, two CVT animal models were used to simulate superior sagittal sinus thrombosis and cortical vein thrombosis. The effects of brain tissue infiltration of NETs and the molecular mechanisms associated with NET formation were deeply explored in combination with proteomics, histology, and serology. The results showed that the cortical vein thrombosis model could be combined with more severe blood-brain barrier (BBB) disruption and showed more severe cerebral hemorrhage. Decreased Sirtuin 1 (SIRT1) expression promotes high mobility group box 1 (HMGB1) acetylation, causing increased cytosolic translocation and extracellular release, and HMGB1 can promote NET formation and recruitment. In addition, corticocerebral accumulation of NETs contributes to BBB damage. This establishes a vicious cycle between BBB damage and NET accumulation. SIRT1 mediated-HMGB1 deacetylation may play a critical role in attenuating BBB damage following CVT. This study employed a combined validation using models of venous sinus thrombosis and cortical vein thrombosis to investigate the deacetylation role of SIRT1, aiming to offer new insights into the pathological mechanisms of brain injury following CVT.
Collapse
Affiliation(s)
- Shuwen Mu
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Ziqi Li
- Department of Neurosurgery, School of Medicine, Dongfang Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361005, China
| | - Long Lin
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Di Wang
- Department of Molecular Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, China
| | - Fei Yang
- Department of Anesthesiology and Perioperative Medicine, 900th Hospital, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Li Chen
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Liang Xian
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Kunzhe Lin
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Yinghong Lin
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Dan Ye
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Yang Yang
- Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Liangfeng Wei
- Department of Neurosurgery, 900th Hospital, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China
| | - Yongjun Xu
- Laboratory of Basic Medicine, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
- Laboratory of Basic Medicine, 900th Hospital, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
| | - Shousen Wang
- Department of Neurosurgery, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
- Department of Neurosurgery, 900th Hospital, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, China.
| |
Collapse
|
34
|
Jiang J, Sun M, Wang Y, Huang W, Xia L. Deciphering the roles of the HMGB family in cancer: Insights from subcellular localization dynamics. Cytokine Growth Factor Rev 2024; 78:85-104. [PMID: 39019664 DOI: 10.1016/j.cytogfr.2024.07.004] [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: 06/03/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
The high-mobility group box (HMGB) family consists of four DNA-binding proteins that regulate chromatin structure and function. In addition to their intracellular functions, recent studies have revealed their involvement as extracellular damage-associated molecular patterns (DAMPs), contributing to immune responses and tumor development. The HMGB family promotes tumorigenesis by modulating multiple processes including proliferation, metabolic reprogramming, metastasis, immune evasion, and drug resistance. Due to the predominant focus on HMGB1 in the literature, little is known about the remaining members of this family. This review summarizes the structural, distributional, as well as functional similarities and distinctions among members of the HMGB family, followed by a comprehensive exploration of their roles in tumor development. We emphasize the distributional and functional hierarchy of the HMGB family at both the organizational and subcellular levels, with a focus on their relationship with the tumor immune microenvironment (TIME), aiming to prospect potential strategies for anticancer therapy.
Collapse
Affiliation(s)
- Junqing Jiang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China
| | - Mengyu Sun
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China
| | - Yufei Wang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province 430030, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi' an 710032, China.
| |
Collapse
|
35
|
Cole-Skinner B, Andre NM, Blankenheim Z, Root KM, Jafri K, Simmons GE. Oleate alters the immune response in non-small cell lung adenocarcinoma through regulation of HMGB1 release. Front Cell Dev Biol 2024; 12:1348707. [PMID: 39100092 PMCID: PMC11294209 DOI: 10.3389/fcell.2024.1348707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 07/10/2024] [Indexed: 08/06/2024] Open
Abstract
Background: Cancer cell evasion of the immune response is critical to cancer development and metastases. Clinicians' ability to kickstart the immune system to target these rogue cells is an ever-growing area of research and medicine. This study delved into the relationship between lipid metabolism, High Mobility Group Box 1 protein (HMGB1)-a pro-inflammatory damage-associated molecular pattern protein-and immune regulation within non-small cell lung adenocarcinoma (NSCLC). Method: To address this question, we used a combination of proteomics, molecular biology, and bioinformatic techniques to investigate the relationship between fatty acids and immune signals within NSCLC. Results: We found that the expression of stearoyl CoA desaturase 1 (SCD1) was decreased in NSCLC tumors compared to normal tissues. This emphasized the critical role of lipid metabolism in tumor progression. Interestingly, monounsaturated fatty acid (MUFA) availability affected the expression of programmed death ligand-1 (PD-L1), a pivotal immune checkpoint target in lung cancer cells and immune cells, as well as HMGB1, suggesting a novel approach to modulating the immune response. This study uncovered a complex interplay between SCD1, PD-L1, and HMGB1, influencing the immunological sensitivity of tumors. Conclusion: Our work underscores the critical importance of understanding the intricate relationships between lipid metabolism and immune modulation to develop more effective NSCLC treatments and personalized therapies. As we continue to explore these connections, we hope to contribute significantly to the ever-evolving field of cancer research, improving patient outcomes and advancing precision medicine in NSCLC.
Collapse
Affiliation(s)
- Breanna Cole-Skinner
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, United States
| | - Nicole M. Andre
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, United States
| | - Zachary Blankenheim
- Department of Biomedical Sciences, School of Medicine, University of Minnesota, Duluth, United States
| | - Kate M. Root
- Department of Biomedical Sciences, School of Medicine, University of Minnesota, Duluth, United States
| | - Kisa Jafri
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, United States
| | - Glenn E. Simmons
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, United States
| |
Collapse
|
36
|
Wang X, Holthauzen LMF, Paz-Villatoro JM, Bien KG, Yu B, Iwahara J. Phosphorylation by Protein Kinase C Weakens DNA-Binding Affinity and Folding Stability of the HMGB1 Protein. Biochemistry 2024; 63:1718-1722. [PMID: 38916994 PMCID: PMC11282465 DOI: 10.1021/acs.biochem.4c00194] [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] [Indexed: 06/27/2024]
Abstract
The HMGB1 protein typically serves as a DNA chaperone that assists DNA-repair enzymes and transcription factors but can translocate from the nucleus to the cytoplasm or even to extracellular space upon some cellular stimuli. One of the factors that triggers the translocation of HMGB1 is its phosphorylation near a nuclear localization sequence by protein kinase C (PKC), although the exact modification sites on HMGB1 remain ambiguous. In this study, using spectroscopic methods, we investigated the HMGB1 phosphorylation and its impact on the molecular properties of the HMGB1 protein. Our nuclear magnetic resonance (NMR) data on the full-length HMGB1 protein showed that PKC specifically phosphorylates the A-box domain, one of the DNA binding domains of HMGB1. Phosphorylation of S46 and S53 was particularly efficient. Over a longer reaction time, PKC phosphorylated some additional residues within the HMGB1 A-box domain. Our fluorescence-based binding assays showed that the phosphorylation significantly reduces the binding affinity of HMGB1 for DNA. Based on the crystal structures of HMGB1-DNA complexes, this effect can be ascribed to electrostatic repulsion between the negatively charged phosphate groups at the S46 side chain and DNA backbone. Our data also showed that the phosphorylation destabilizes the folding of the A-box domain. Thus, phosphorylation by PKC weakens the DNA-binding affinity and folding stability of HMGB1.
Collapse
Affiliation(s)
- Xi Wang
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| | - Luis Marcelo F. Holthauzen
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| | - Jonathan M Paz-Villatoro
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| | - Karina G. Bien
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| | - Binhan Yu
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| | - Junji Iwahara
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, Texas 77555-1068, USA
| |
Collapse
|
37
|
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
|
38
|
Chikhirzhina E, Tsimokha A, Tomilin AN, Polyanichko A. Structure and Functions of HMGB3 Protein. Int J Mol Sci 2024; 25:7656. [PMID: 39062899 PMCID: PMC11276821 DOI: 10.3390/ijms25147656] [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: 05/24/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
HMGB3 protein belongs to the group of HMGB proteins from the superfamily of nuclear proteins with high electrophoretic mobility. HMGB proteins play an active part in almost all cellular processes associated with DNA-repair, replication, recombination, and transcription-and, additionally, can act as cytokines during infectious processes, inflammatory responses, and injuries. Although the structure and functions of HMGB1 and HMGB2 proteins have been intensively studied for decades, very little attention has been paid to HMGB3 until recently. In this review, we summarize the currently available data on the molecular structure, post-translational modifications, and biological functions of HMGB3, as well as the possible role of the ubiquitin-proteasome system-dependent HMGB3 degradation in tumor development.
Collapse
Affiliation(s)
- Elena Chikhirzhina
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Av. 4, 194064 St. Petersburg, Russia; (A.T.); (A.N.T.); (A.P.)
| | | | | | | |
Collapse
|
39
|
Mo C, Huang Q, Li L, Long Y, Shi Y, Lu Z, Wu N, Li Q, Zeng H, Li G, Qiu L, Gui C, Ji Q. High-mobility group box 1 and its related receptors: potential therapeutic targets for contrast-induced acute kidney injury. Int Urol Nephrol 2024; 56:2291-2299. [PMID: 38438703 DOI: 10.1007/s11255-024-03981-2] [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: 12/04/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024]
Abstract
Percutaneous coronary intervention (PCI) is a crucial diagnostic and therapeutic approach for coronary heart disease. Contrast agents' exposure during PCI is associated with a risk of contrast-induced acute kidney injury (CI-AKI). CI-AKI is characterized by a sudden decline in renal function occurring as a result of exposure to intravascular contrast agents, which is associated with an increased risk of poor prognosis. The pathophysiological mechanisms underlying CI-AKI involve renal medullary hypoxia, direct cytotoxic effects, endoplasmic reticulum stress, inflammation, oxidative stress, and apoptosis. To date, there is no effective therapy for CI-AKI. High-mobility group box 1 (HMGB1), as a damage-associated molecular pattern molecule, is released extracellularly by damaged cells or activated immune cells and binds to related receptors, including toll-like receptors and receptor for advanced glycation end product. In renal injury, HMGB1 is expressed in renal tubular epithelial cells, macrophages, endothelial cells, and glomerular cells, involved in the pathogenesis of various kidney diseases by activating its receptors. Therefore, this review provides a theoretical basis for HMGB1 as a therapeutic intervention target for CI-AKI.
Collapse
Affiliation(s)
- Changhua Mo
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qili Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Lixia Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yusheng Long
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Zhengde Lu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Ning Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qingkuan Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Huayuan Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Guihua Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Lingyue Qiu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University and Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention and Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning, China.
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China.
| |
Collapse
|
40
|
Takahata S, Taguchi A, Takenaka A, Mori M, Chikashige Y, Tsutsumi C, Hiraoka Y, Murakami Y. The HMG-box module in FACT is critical for suppressing epigenetic variegation of heterochromatin in fission yeast. Genes Cells 2024; 29:567-583. [PMID: 38837646 DOI: 10.1111/gtc.13132] [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: 03/26/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 06/07/2024]
Abstract
Chromatin condensation state is the key for retrieving genetic information. High-mobility group protein (HMG) proteins exhibit DNA-binding and bending activities, playing an important role in the regulation of chromatin structure. We have shown that nucleosomes tightly packaged into heterochromatin undergo considerable dynamic histone H2A-H2B maintenance via the direct interaction between HP1/Swi6 and facilitate chromatin transcription (FACT), which is composed of the Spt16/Pob3 heterodimer and Nhp6. In this study, we analyzed the role of Nhp6, an HMG box protein, in the FACT at heterochromatin. Pob3 mutant strains showed derepressed heterochromatin-dependent gene silencing, whereas Nhp6 mutant strains did not show significant defects in chromatin regulation or gene expression, suggesting that these two modules play different roles in chromatin regulation. We expressed a protein fusing Nhp6 to the C-terminus of Pob3, which mimics the multicellular FACT component Ssrp1. The chromatin-binding activity of FACT increased with the number of Nhp6 fused to Pob3, and the heterochromatin formation rate was promoted more strongly. Furthermore, we demonstrated that this promotion of heterochromatinization inhibited the heterochromatic variegation caused by epe1+ disruption. Heterochromatic variegation can be observed in a variety of regulatory steps; however, when it is caused by fluctuations in chromatin arrangement, it can be eliminated through the strong recruitment of the FACT complex.
Collapse
Affiliation(s)
- Shinya Takahata
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Asahi Taguchi
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo, Japan
| | - Ayaka Takenaka
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo, Japan
| | - Miyuki Mori
- Graduate School of Chemical Sciences and Engineering, Hokkaido University, Sapporo, Japan
| | - Yuji Chikashige
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Chihiro Tsutsumi
- Advanced ICT Research Institute, National Institute of Information and Communications Technology, Kobe, Japan
| | - Yasushi Hiraoka
- Graduate School of Frontier Biosciences, Osaka University, Suita, Japan
| | - Yota Murakami
- Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo, Japan
| |
Collapse
|
41
|
(Ogi) Suzuki K, Okamoto T, Tamai K, Tabata Y, Hatano E. Enhancement of tracheal cartilage regeneration by local controlled release of stromal cell-derived factor 1α with gelatin hydrogels and systemic administration of high-mobility group box 1 peptide. Regen Ther 2024; 26:415-424. [PMID: 39070123 PMCID: PMC11282968 DOI: 10.1016/j.reth.2024.06.017] [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: 04/01/2024] [Revised: 06/19/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Introduction This present study evaluated the effect of combination therapy with stromal cell-derived factor 1α (SDF-1α) and high-mobility group box 1 (HMGB1) peptide on the regeneration of tracheal injury in a rat model. Methods To improve this effect, SDF-1α was incorporated into a gelatin hydrogel, which was then applied to the damaged tracheal cartilage of rats for local release. Furthermore, HMGB1 peptide was repeatedly administered intravenously. Regeneration of damaged tracheal cartilage was evaluated in terms of cell recruitment. Results Mesenchymal stem cells (MSC) with C-X-C motif chemokine receptor 4 (CXCR4) were mobilized more into the injured area, and consequently the fastest tracheal cartilage regeneration was observed in the combination therapy group eight weeks after injury. Conclusions The present study demonstrated that combination therapy with gelatin hydrogel incorporating SDF-1α and HMGB1 peptide injected intravenously can enhance the recruitment of CXCR4-positive MSC, promoting the regeneration of damaged tracheal cartilage.
Collapse
Affiliation(s)
- Kumiko (Ogi) Suzuki
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Biomaterials, Field of Tissue Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tatsuya Okamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Katsuto Tamai
- Department of Stem Cell Therapy Science, Graduate School of Medicine, Osaka University, Japan
| | - Yasuhiko Tabata
- Department of Biomaterials, Field of Tissue Engineering, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Etsuro Hatano
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
42
|
Patra S, Roy PK, Dey A, Mandal M. Impact of HMGB1 on cancer development and therapeutic insights focused on CNS malignancy. Biochim Biophys Acta Rev Cancer 2024; 1879:189105. [PMID: 38701938 DOI: 10.1016/j.bbcan.2024.189105] [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: 02/07/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
The present study explores the complex roles of High Mobility Group Box 1 (HMGB1) in the context of cancer development, emphasizing glioblastoma (GBM) and other central nervous system (CNS) cancers. HMGB1, primarily known for its involvement in inflammation and angiogenesis, emerges as a multifaceted player in the tumorigenesis of GBM. The overexpression of HMGB1 correlates with glioma malignancy, influencing key pathways like RAGE/MEK/ERK and RAGE/Rac1. Additionally, HMGB1 secretion is linked to the maintenance of glioma stem cells (GSCs) and contributes to the tumor microenvironment's (TME) vascular leakiness. Henceforth, our review discusses the bidirectional impact of HMGB1, acting as both a promoter of tumor progression and a mediator of anti-tumor immune responses. Notably, HMGB1 exhibits tumor-suppressive roles by inducing apoptosis, limiting cellular proliferation, and enhancing the sensitivity of GBM to therapeutic interventions. This dualistic nature of HMGB1 calls for a nuanced understanding of its implications in GBM pathogenesis, offering potential avenues for more effective and personalized treatment strategies. The findings underscore the need to explore HMGB1 as a prognostic marker, therapeutic target, and a promising tool for stimulating anti-tumor immunity in GBM.
Collapse
Affiliation(s)
- Sucharita Patra
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| | - Pritam Kumar Roy
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| | - Ankita Dey
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| | - Mahitosh Mandal
- Cancer Biology Lab, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal, India.
| |
Collapse
|
43
|
Lv G, Yang M, Gai K, Jia Q, Wang Z, Wang B, Li X. Multiple functions of HMGB1 in cancer. Front Oncol 2024; 14:1384109. [PMID: 38725632 PMCID: PMC11079206 DOI: 10.3389/fonc.2024.1384109] [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: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
High mobility group box 1 (HMGB1) is a nuclear DNA-binding protein with a dual role in cancer, acting as an oncogene and a tumor suppressor. This protein regulates nucleosomal structure, DNA damage repair, and genomic stability within the cell, while also playing a role in immune cell functions. This review comprehensively evaluates the biological and clinical significance of HMGB1 in cancer, including its involvement in cell death and survival, its potential as a therapeutic target and cancer biomarker, and as a prosurvival signal for the remaining cells after exposure to cytotoxic anticancer treatments. We highlight the need for a better understanding of the cellular markers and mechanisms involved in the involvement of HMGB1in cancer, and aim to provide a deeper understanding of its role in cancer progression.
Collapse
Affiliation(s)
- Guangyao Lv
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Menglin Yang
- Quality Management Department, Marine Biomedical Research Institute of Qingdao, Qingdao, China
| | - Keke Gai
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Qiong Jia
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Zhenzhen Wang
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Bin Wang
- Department of Pharmacy, Binzhou Medical University Hospital, Binzhou, Shandong, China
| | - Xueying Li
- School of Health, Binzhou Polytechnic, Binzhou, China
| |
Collapse
|
44
|
Liao M, Cao J, Chen W, Wang M, Jin Z, Ye J, Ren Y, Wei Y, Xue Y, Chen D, Zhang Y, Chen S. HMGB1 prefers to interact with structural RNAs and regulates rRNA methylation modification and translation in HeLa cells. BMC Genomics 2024; 25:345. [PMID: 38580917 PMCID: PMC10996203 DOI: 10.1186/s12864-024-10204-6] [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: 05/30/2023] [Accepted: 03/08/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND High-mobility group B1 (HMGB1) is both a DNA binding nuclear factor modulating transcription and a crucial cytokine that mediates the response to both infectious and noninfectious inflammation such as autoimmunity, cancer, trauma, and ischemia reperfusion injury. HMGB1 has been proposed to control ribosome biogenesis, similar as the other members of a class of HMGB proteins. RESULTS Here, we report that HMGB1 selectively promotes transcription of genes involved in the regulation of transcription, osteoclast differentiation and apoptotic process. Improved RNA immunoprecipitation by UV cross-linking and deep sequencing (iRIP-seq) experiment revealed that HMGB1 selectively bound to mRNAs functioning not only in signal transduction and gene expression, but also in axon guidance, focal adhesion, and extracellular matrix organization. Importantly, HMGB1-bound reads were strongly enriched in specific structured RNAs, including the domain II of 28S rRNA, H/ACA box snoRNAs including snoRNA63 and scaRNAs. RTL-P experiment showed that overexpression of HMGB1 led to a decreased methylation modification of 28S rRNA at position Am2388, Cm2409, and Gm2411. We further showed that HMGB1 overexpression increased ribosome RNA expression levels and enhanced protein synthesis. CONCLUSION Taken together, our results support a model in which HMGB1 binds to multiple RNA species in human cancer cells, which could at least partially contribute to HMGB1-modulated rRNA modification, protein synthesis function of ribosomes, and differential gene expression including rRNA genes. These findings provide additional mechanistic clues to HMGB1 functions in cancers and cell differentiation.
Collapse
Affiliation(s)
- Meimei Liao
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Jiarui Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Wen Chen
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Mengwei Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Zhihui Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Jia Ye
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Yijun Ren
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China
| | - Yaxun Wei
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Yaqiang Xue
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Dong Chen
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
- Laboratory for Genome Regulation and Human Health, ABLife Inc., Optics Valley International Biomedical Park, East Lake High-Tech Development Zone, 388 Gaoxin 2Nd Road, Hubei, Wuhan, 430075, China
| | - Sen Chen
- Department of Orthopedics, Renmin Hospital of Wuhan University, Hubei, Wuhan, People's Republic of China.
| |
Collapse
|
45
|
Park YJ, Heo JB, Choi YJ, Cho S, Lee T, Song GY, Bae JS. Antiseptic Functions of CGK012 against HMGB1-Mediated Septic Responses. Int J Mol Sci 2024; 25:2976. [PMID: 38474222 PMCID: PMC10931621 DOI: 10.3390/ijms25052976] [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: 01/31/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
High mobility group box 1 (HMGB1), a protein with important functions, has been recognized as a potential therapeutic target for the treatment of sepsis. One possible mechanism for this is that inhibiting HMGB1 secretion can exert antiseptic effects, which can restore the integrity of the vascular barrier. (7S)-(+)-cyclopentyl carbamic acid 8,8-dimethyl-2-oxo-6,7-dihydro-2H,8H-pyrano[3,2-g]chromen-7-yl-ester (CGK012) is a newly synthesized pyranocoumarin compound that could function as a novel small-molecule inhibitor of the Wnt/β-catenin signaling pathway. However, no studies have yet determined the effects of CGK012 on sepsis. We investigated the potential of CGK012 to attenuate the excessive permeability induced by HMGB1 and enhance survival rates in a mouse model of sepsis with reduced HMGB1 levels following lipopolysaccharide (LPS) treatment. In both LPS-stimulated human endothelial cells and a mouse model exhibiting septic symptoms due to cecal ligation and puncture (CLP), we assessed proinflammatory protein levels and tissue damage biomarkers as indicators of reduced vascular permeability. CGK012 was applied after induction in human endothelial cells exposed to LPS and the CLP-induced mouse model of sepsis. CGK012 effectively mitigated excessive permeability and suppressed HMGB1 release, resulting in improved vascular stability, decreased mortality, and enhanced histological conditions in the mouse model of CLP-induced sepsis. In conclusion, our findings indicate that CGK012 treatment in mice with CLP-induced sepsis diminished HMGB1 release and increased the survival rate, suggesting its potential as a pharmaceutical intervention for sepsis.
Collapse
Affiliation(s)
- Yun Jin Park
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (Y.J.P.); (S.C.); (T.L.)
| | - Jong Beom Heo
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea; (J.B.H.); (Y.-J.C.)
| | - Yoon-Jung Choi
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea; (J.B.H.); (Y.-J.C.)
| | - Sanghee Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (Y.J.P.); (S.C.); (T.L.)
| | - Taeho Lee
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (Y.J.P.); (S.C.); (T.L.)
| | - Gyu Yong Song
- College of Pharmacy, Chungnam National University, Daejon 34134, Republic of Korea; (J.B.H.); (Y.-J.C.)
- AREZ Co., Ltd., Daejeon 34036, Republic of Korea
| | - Jong-Sup Bae
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Kyungpook National University, Daegu 41566, Republic of Korea; (Y.J.P.); (S.C.); (T.L.)
| |
Collapse
|
46
|
Vásquez-Suárez A, Muñoz-Flores C, Ortega L, Roa F, Castillo C, Romero A, Parra N, Sandoval F, Macaya L, González-Chavarría I, Astuya A, Starck MF, Villegas MF, Agurto N, Montesino R, Sánchez O, Valenzuela A, Toledo JR, Acosta J. Design and functional characterization of Salmo salar TLR5 agonist peptides derived from high mobility group B1 acidic tail. FISH & SHELLFISH IMMUNOLOGY 2024; 146:109373. [PMID: 38272332 DOI: 10.1016/j.fsi.2024.109373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Toll-like receptor 5 (TLR5) responds to the monomeric form of flagellin and induces the MyD88-depending signaling pathway, activating proinflammatory transcription factors such as NF-κB and the consequent induction of cytokines. On the other hand, HMGB1 is a highly conserved non-histone chromosomal protein shown to interact with and activate TLR5. The present work aimed to design and characterize TLR5 agonist peptides derived from the acidic tail of Salmo salar HMGB1 based on the structural knowledge of the TLR5 surface using global molecular docking platforms. Peptide binding poses complexed on TLR5 ectodomain model from each algorithm were filtrated based on docking scoring functions and predicted theoretical binding affinity of the complex. Circular dichroism spectra were recorded for each peptide selected for synthesis. Only intrinsically disordered peptides (6W, 11W, and SsOri) were selected for experimental functional assay. The functional characterization of the peptides was performed by NF-κB activation assays, RT-qPCR gene expression assays, and Piscirickettsia salmonis challenge in SHK-1 cells. The 6W and 11W peptides increased the nuclear translation of p65 and phosphorylation. In addition, the peptides induced the expression of genes related to the TLR5 pathway activation, pro- and anti-inflammatory response, and differentiation and activation of T lymphocytes towards phenotypes such as TH1, TH17, and TH2. Finally, it was shown that the 11W peptide protects immune cells against infection with P. salmonis bacteria. Overall, the results indicate the usefulness of novel peptides as potential immunostimulants in salmonids.
Collapse
Affiliation(s)
- Aleikar Vásquez-Suárez
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carolina Muñoz-Flores
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Leonardo Ortega
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Francisco Roa
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Carolina Castillo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Alex Romero
- Laboratorio de Inmunología y Estrés de Organismos Acuáticos, Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile; Centro FONDAP, Interdisciplinary Center for Aquaculture Research (INCAR), Universidad de Concepción, Concepción, Chile
| | - Natalie Parra
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Felipe Sandoval
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Luis Macaya
- Departamento de Físico-Química, Facultad de Ciencias Químicas, Universidad de Concepción, Concepción, Chile
| | - Iván González-Chavarría
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Allisson Astuya
- Laboratorio de Genómica Marina y Cultivo Celular, Departamento de Oceanografía y COPAS Sur-Austral, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - María Francisca Starck
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Milton F Villegas
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Niza Agurto
- Laboratorio de Piscicultura y Patología Acuática, Departamento de Oceanografía, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Raquel Montesino
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Oliberto Sánchez
- Laboratorio de Biofármacos Recombinantes, Departamento de Farmacología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Ariel Valenzuela
- Laboratorio de Piscicultura y Patología Acuática, Departamento de Oceanografía, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Jorge R Toledo
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - Jannel Acosta
- Laboratorio de Biotecnología y Biofármacos, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| |
Collapse
|
47
|
Sakata N. The anti-inflammatory effect of metformin: The molecular targets. Genes Cells 2024; 29:183-191. [PMID: 38311861 PMCID: PMC11448366 DOI: 10.1111/gtc.13098] [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: 09/28/2023] [Revised: 12/24/2023] [Accepted: 01/11/2024] [Indexed: 02/06/2024]
Abstract
Metformin is an anti-diabetic drug. Metformin mainly inhibits gluconeogenesis in the liver and reduces blood sugar. In addition to the anti-diabetic effects, many studies have revealed that metformin has anti-inflammatory effects. Various molecules were suggested to be the target of the metformin's anti-inflammatory effects. However, the conclusion is not clear. Metformin is related to a number of molecules and the identification of the main target in anti-inflammatory effects leads to the understanding of inflammation and metformin. In this article, I discuss each suggested molecule, involved mechanisms, and their relationship with various diseases.
Collapse
|
48
|
Meng X, Na R, Peng X, Li H, Ouyang W, Zhou W, You X, Li Y, Pu X, Zhang K, Xia J, Wang J, Tang H, Zhuang G, Peng Z. Musashi-2 potentiates colorectal cancer immune infiltration by regulating the post-translational modifications of HMGB1 to promote DCs maturation and migration. Cell Commun Signal 2024; 22:117. [PMID: 38347600 PMCID: PMC10863188 DOI: 10.1186/s12964-024-01495-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/21/2024] [Indexed: 02/15/2024] Open
Abstract
Post-translational modifications (PTMs) of the non-histone protein high-mobility group protein B1 (HMGB1) are involved in modulating inflammation and immune responses. Recent studies have implicated that the RNA-binding protein (RBP) Musashi-2 (MSI2) regulates multiple critical biological metabolic and immunoregulatory functions. However, the precise role of MSI2 in regulating PTMs and tumor immunity in colorectal cancer (CRC) remains unclear. Here, we present data indicating that MSI2 potentiates CRC immunopathology in colitis-associated colon cancer (CAC) mouse models, cell lines and clinical specimens, specifically via HMGB1-mediated dendritic cell (DC) maturation and migration, further contributes to the infiltration of CD4+ and CD8+ T cells and inflammatory responses. Under stress conditions, MSI2 can exacerbate the production, nucleocytoplasmic transport and extracellular release of damage-associated molecular patterns (DAMPs)-HMGB1 in CRC cells. Mechanistically, MSI2 mainly enhances the disulfide HMGB1 production and protein translation via direct binding to nucleotides 1403-1409 in the HMGB1 3' UTR, and interacts with the cytoplasmic acetyltransferase P300 to upregulate its expression, further promoting the acetylation of K29 residue in HMGB1, thus leading to K29-HMGB1 nucleocytoplasmic translocation and extracellular release. Furthermore, blocking HMGB1 activity with glycyrrhizic acid (Gly) attenuates MSI2-mediated immunopathology and immune infiltration in CRC in vitro and in vivo. Collectively, this study suggests that MSI2 may improve the prognosis of CRC patients by reprogramming the tumor immune microenvironment (TIME) through HMGB1-mediated PTMs, which might be a novel therapeutic option for CRC immunotherapy.
Collapse
Affiliation(s)
- Xiaole Meng
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Xiamen Clinical Research Center for Cancer Therapy; Department of Pathology, Zhongshan Hospital (Xiamen Branch), Fudan University; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Risi Na
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xiao Peng
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Hui Li
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wanxin Ouyang
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Wenting Zhou
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xuting You
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yuhuan Li
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xin Pu
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Ke Zhang
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Junjie Xia
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jie Wang
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Huamei Tang
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Department of Pathology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Guohong Zhuang
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Zhihai Peng
- Organ Transplantation Institute of Xiamen University; Xiamen Human Organ Transplantation Quality Control Center; Xiamen Key Laboratory of Regeneration Medicine; Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
- Organ Transplantation Clinical Medical Center of Xiamen University; Department of General Surgery, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
49
|
Awad AM, Elshaer SL, Gangaraju R, Abdelaziz RR, Nader MA. Ameliorative effect of montelukast against STZ induced diabetic nephropathy: targeting HMGB1, TLR4, NF-κB, NLRP3 inflammasome, and autophagy pathways. Inflammopharmacology 2024; 32:495-508. [PMID: 37498374 PMCID: PMC10907471 DOI: 10.1007/s10787-023-01301-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/04/2023] [Indexed: 07/28/2023]
Abstract
Diabetic nephropathy (DN) is reported as one of the most serious microvascular diabetic complications and the trigger of end-stage renal disease (ESRD), underscoring the concern of any therapeutic intervention directed at ameliorating the development and progression of DN. The current study explored the renoprotective impact of montelukast (Mon) against streptozotocin (STZ)-induced DN in rats compared to a standard anti-hyperglycemic insulin (Ins) treatment. Diabetes was induced by a single dose of STZ (55 mg/kg). Diabetic rats were treated with Mon (10 and 20 mg/kg, oral gavage) for eight weeks. Mon administration for 8 weeks after induction of diabetes conferred significant dose-dependent renoprotection, independent of blood glucose levels (unlike Ins), as evidenced by the improvement in serum creatinine, and blood urea nitrogen (BUN), and ameliorated STZ-induced renal necrotic, inflammatory alterations, and renal fibrosis. Additionally, Mon treatment in diabetic rats significantly restored redox hemostasis as evidenced by malondialdehyde (MDA) and total antioxidant capacity (TAC) levels; significantly reduced the renal expression of high mobility group box (HMGB) 1, toll-like receptor (TLR) 4, nuclear factor kappa B (NF-κB) (in the nucleus), NOD-like receptor family pyrin domain containing (NLRP) 3, and interleukin (IL)-1β. Moreover, Mon administration ameliorated the dysregulation in autophagy as evidenced by p62 and microtubule-associated protein 1A/1B-light chain 3 (LC3)-II levels. In conclusion, the renoprotective effect of Mon is potentially associated with its modulatory effect on inflammatory cytokines, antioxidant properties, and autophagy.
Collapse
Affiliation(s)
- Ahmed M Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Sally L Elshaer
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
50
|
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
|