1
|
Jang S, Lee JB, Yoo C, Kim HS, Choi K, Lee J, Lee DY. Biocompatible and nondegradable microcapsules using an ethylamine-bridged EGCG dimer for successful therapeutic cell transplantation. J Control Release 2024; 373:520-532. [PMID: 39059498 DOI: 10.1016/j.jconrel.2024.07.053] [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: 02/23/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Conventional alginate microcapsules are widely used for encapsulating therapeutic cells to reduce the host immune response. However, the exchange of monovalent cations with divalent cations for crosslinking can lead to a sol-gel phase transition, resulting in gradual degradation and swelling of the microcapsules in the body. To address this limitation, we present a biocompatible and nondegradable epigallocatechin-3-gallate (EGCG)-based microencapsulation with ethylamine-bridged EGCG dimers (EGCG(d)), denoted as 'Epi-Capsules'. These Epi-Capsules showed increased physical properties and Ca2+ chelating resistance compared to conventional alginate microcapsules. Horseradish peroxidase (HRP) treatment is very effective in increasing the stability of Epi-Capsule((+)HRP) due to the crosslinking between EGCG(d) molecules. Interestingly, the Epi-Capsules(oxi) using a pre-oxidized EGCG(d) can support long-term survival (>90 days) of xenotransplanted insulin-secreting islets in diabetic mice in vivo, which is attributed to its structural stability and reactive oxygen species (ROS) scavenging for lower fibrotic activity. Collectively, this EGCG-based microencapsulation can create Ca2+ chelating-resistance and anti-oxidant activity, which could be a promising strategy for cell therapies for diabetes and other diseases.
Collapse
Affiliation(s)
- Seonmi Jang
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Jae Bin Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Chaerim Yoo
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyung Shik Kim
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea
| | - Kimyung Choi
- Optipharm Co., Ltd., Cheongju 28158, Republic of Korea
| | - Joonseok Lee
- Department of Chemistry, Hanyang University, Seoul 04763, Republic of Korea.
| | - Dong Yun Lee
- Department of Bioengineering, College of Engineering, and BK FOUR Biopharmaceutical Innovation Leader for Education and Research Group, Hanyang University, Seoul 04763, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul 04763, Republic of Korea; Institute for Bioengineering and Biopharmaceutical Research (IBBR), Hanyang University, Seoul 04763, Republic of Korea; Elixir Pharmatech Inc., Seoul 04763, Republic of Korea.
| |
Collapse
|
2
|
Wulandari S, Hartono, Wibawa T. The role of HMGB1 in COVID-19-induced cytokine storm and its potential therapeutic targets: A review. Immunology 2023; 169:117-131. [PMID: 36571562 PMCID: PMC9880760 DOI: 10.1111/imm.13623] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/22/2022] [Indexed: 12/27/2022] Open
Abstract
Hyperinflammation characterized by elevated proinflammatory cytokines known as 'cytokine storms' is the major cause of high severity and mortality seen in COVID-19 patients. The pathology behind the cytokine storms is currently unknown. Increased HMGB1 levels in serum/plasma of COVID-19 patients were reported by many studies, which positively correlated with the level of proinflammatory cytokines. Dead cells following SARS-CoV-2 infection might release a large amount of HMGB1 and RNA of SARS-CoV-2 into extracellular space. HMGB1 is a well-known inflammatory mediator. Additionally, extracellular HMGB1 might interact with SARS-CoV-2 RNA because of its high capability to bind with a wide variety of molecules including nucleic acids and could trigger massive proinflammatory immune responses. This review aimed to critically explore the many possible pathways by which HMGB1-SARS-CoV-2 RNA complexes mediate proinflammatory responses in COVID-19. The contribution of these pathways to impair host immune responses against SARS-CoV-2 infection leading to a cytokine storm was also evaluated. Moreover, since blocking the HMGB1-SARS-CoV-2 RNA interaction might have therapeutic value, some of the HMGB1 antagonists have been reviewed. The HMGB1- SARS-CoV-2 RNA complexes might trigger endocytosis via RAGE which is linked to lysosomal rupture, PRRs activation, and pyroptotic death. High levels of the proinflammatory cytokines produced might suppress many immune cells leading to uncontrolled viral infection and cell damage with more HMGB1 released. Altogether these mechanisms might initiate a proinflammatory cycle leading to a cytokine storm. HMGB1 antagonists could be considered to give benefit in alleviating cytokine storms and serve as a potential candidate for COVID-19 therapy.
Collapse
Affiliation(s)
- Sri Wulandari
- Doctorate Program of Medicine and Health Science, Faculty of MedicinePublic Health and Nursing Universitas Gadjah MadaYogyakartaIndonesia
- Department of Physiology, Faculty of MedicineUniversitas Sebelas MaretSurakartaIndonesia
| | - Hartono
- Department of Physiology, Faculty of MedicineUniversitas Sebelas MaretSurakartaIndonesia
| | - Tri Wibawa
- Department of Microbiology, Faculty of MedicinePublic Health and Nursing Universitas Gadjah MadaYogyakartaIndonesia
| |
Collapse
|
3
|
Chen Y, Liu Z, Gong Y. Neuron-immunity communication: mechanism of neuroprotective effects in EGCG. Crit Rev Food Sci Nutr 2023; 64:9333-9352. [PMID: 37216484 DOI: 10.1080/10408398.2023.2212069] [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] [Indexed: 05/24/2023]
Abstract
Epigallocatechin gallate (EGCG), a naturally occurring active ingredient unique to tea, has been shown to have neuroprotective potential. There is growing evidence of its potential advantages in the prevention and treatment of neuroinflammation, neurodegenerative diseases, and neurological damage. Neuroimmune communication is an important physiological mechanism in neurological diseases, including immune cell activation and response, cytokine delivery. EGCG shows great neuroprotective potential by modulating signals related to autoimmune response and improving communication between the nervous system and the immune system, effectively reducing the inflammatory state and neurological function. During neuroimmune communication, EGCG promotes the secretion of neurotrophic factors into the repair of damaged neurons, improves intestinal microenvironmental homeostasis, and ameliorates pathological phenotypes through molecular and cellular mechanisms related to the brain-gut axis. Here, we discuss the molecular and cellular mechanisms of inflammatory signaling exchange involving neuroimmune communication. We further emphasize that the neuroprotective role of EGCG is dependent on the modulatory role between immunity and neurology in neurologically related diseases.
Collapse
Affiliation(s)
- Ying Chen
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
- Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Changsha, China
- Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha, China
| | - Yushun Gong
- Key Laboratory of Tea Science of Ministry of Educatioxn, Changsha, China
- National Research Center of Engineering and Technology for Utilization of Botanical Functional Ingredients, Changsha, China
| |
Collapse
|
4
|
Shen P, Sun Y, Jiang X, Zhou X, Nian B, Wang W, Zhang J. Interaction of bioactive kaempferol with HMGB1: Investigation by multi-spectroscopic and molecular simulation methods. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 292:122360. [PMID: 36724682 DOI: 10.1016/j.saa.2023.122360] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
Chronic and persistent inflammation associated with excessive high mobility group protein 1 (HMGB1) is a risk factor for various diseases. Dietary intake of kaempferol has been proven to be effective in reducing HMGB1 levels and the degree of inflammation, but the structural mechanism remains unclear. In this context, we first investigated the interaction between bioactive kaempferol and HMGB1 using multi-spectroscopic and molecular simulation techniques. The surface plasmon resonance (SPR) data indicated that kaempferol binds directly to HMGB1 with a Kd value of 2.89 × 10-5 M. Binding of kaempferol with HMGB1 led to the intrinsic fluorescence quenching and modest secondary structure change of HMGB1 supported by fluorescence spectrometry and circular dichroism (CD). Using dynamic light scattering (DLS), it was found that kaempferol induced the aggregation of HMGB1 protein complex to form larger particles. On HMGB1-activated RAW264.7 cells, kaempferol co-incubation exhibited a remarkable inhibitory effect on nitric oxide (NO) release with an IC50 value of 5.02 μM, which was lower than that of quercetin. In silico, kaempferol binds to HMGB1 mainly through hydrogen bonds and hydrophobic forces. Collectively, our study showed kaempferol as a potential HMGB1 inhibitor, mainly acting by direct binding to HMGB1 and inducing its conformational changes, which provides clues for the treatment of chronic inflammation by kaempferol.
Collapse
Affiliation(s)
- Pingping Shen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Yueming Sun
- The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou 550025, PR China
| | - Xuewa Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Xiaoyang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China
| | - Binbin Nian
- RWTH Aachen University, Aachen 52062, Germany
| | - Weiwei Wang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing 210046, PR China
| | - Jian Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
5
|
miR-22 alleviates sepsis-induced acute kidney injury via targeting the HMGB1/TLR4/NF-κB signaling pathway. Int Urol Nephrol 2023; 55:409-421. [PMID: 35960478 PMCID: PMC9859886 DOI: 10.1007/s11255-022-03321-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 07/24/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Acute kidney injury (AKI) is a severe complication of sepsis, and is strongly correlated with MicroRNAs (miRNAs). However, the mechanism of miR-22 on sepsis-induced AKI is not clearly understood. The study aimed to explore the role and mechanism of miR-22 on AKI. METHODS The AKI models were established by cecal ligation and puncture (CLP) surgery in SD rats and lipopolysaccharide (LPS) induction in HBZY-1 cells. In AKI rats, the content of serum creatinine (SCr) and blood urea nitrogen (BUN) were detected. Kidney tissues were pathologically examined by H&E and PAS staining. The LPS-induced HBZY-1 cells were transfected with mimics miR-22, si-HMGB1, or oe-HMGB1. miR-22 and HMGB1 expression was detected in vivo and in vitro. In transfected cells, HMGB1/TLR4/NF-κB pathway-related protein expressions were measured by Western blot. The relationship between miR-22 and HMGB1 was assessed by a dual-luciferase gene report. Inflammatory cytokine levels in serum and cells were assessed by ELISA. RESULTS In AKI rats, kidney injury was observed, accompanied by the down-regulated miR-122 expression and up-regulated HMBG1 expression. The dual-luciferase report found miR-22-3p could targetly regulate HMBG1. Furthermore, both in vitro and in vivo experiments revealed that the releases of inflammatory cytokine were increased after AKI modeling, but the situation was reversed by mimics miR-22 or si-HMGB1 in vitro. In HBZY-1 cells, mimics miR-22 could suppress LPS-induced overexpression of HMGB1/TLR4/NF-κB signaling pathway-related proteins. However, the oe-HMGB1 addition reversed the effect of mimics miR-22. CONCLUSION miR-22 can inhibit the inflammatory response, target the HMGB1, and inhibit the HMGB1/TLR4/NF-kB pathway, to attenuate the sepsis-induced AKI, which indicates that miR-22 may serve as a potential treatment target in sepsis-induced AKI.
Collapse
|
6
|
He M, Chu T, Wang Z, Feng Y, Shi R, He M, Feng S, Lu L, Cai C, Fang F, Zhang X, Liu Y, Gao B. Inhibition of macrophages inflammasome activation via autophagic degradation of HMGB1 by EGCG ameliorates HBV-induced liver injury and fibrosis. Front Immunol 2023; 14:1147379. [PMID: 37122751 PMCID: PMC10140519 DOI: 10.3389/fimmu.2023.1147379] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/29/2023] [Indexed: 05/02/2023] Open
Abstract
Background Liver fibrosis is a reversible wound-healing response that can lead to end-stage liver diseases without effective treatment, in which HBV infection is a major cause. However, the underlying mechanisms for the development of HBV-induced fibrosis remains elusive, and efficacious therapies for this disease are still lacking. In present investigation, we investigated the effect and mechanism of green tea polyphenol epigallocatechin-3-gallate (EGCG) on HBV-induced liver injury and fibrosis. Methods The effect of EGCG on liver fibrosis was examined in a recombinant cccDNA (rcccDNA) chronic HBV mouse model by immunohistochemical staining, Sirius red and Masson's trichrome staining. The functional relevance between high mobility group box 1 (HMGB1) and inflammasome activation and the role of EGCG in it were analyzed by Western blotting. The effect of EGCG on autophagic flux was determined by Western blotting and flow cytometric analysis. Results EGCG treatment efficiently was found to alleviate HBV-induced liver injury and fibrosis in a recombinant cccDNA (rcccDNA) chronic HBV mouse model, a proven suitable research platform for HBV-induced fibrosis. Mechanistically, EGCG was revealed to repress the activation of macrophage NLRP3 inflammasome, a critical trigger of HBV-induced liver fibrosis. Further study revealed that EGCG suppressed macrophage inflammasome through downregulating the level of extracellular HMGB1. Furthermore, our data demonstrated that EGCG treatment downregulated the levels of extracellular HMGB1 through activating autophagic degradation of cytoplasmic HMGB1 in hepatocytes. Accordingly, autophagy blockade was revealed to significantly reverse EGCG-mediated inhibition on extracellular HMGB1-activated macrophage inflammasome and thus suppress the therapeutic effect of EGCG on HBV-induced liver injury and fibrosis. Conclusion EGCG ameliorates HBV-induced liver injury and fibrosis via autophagic degradation of cytoplasmic HMGB1 and the subsequent suppression of macrophage inflammasome activation. These data provided a new pathogenic mechanism for HBV-induced liver fibrosis involving the extracellular HMGB1-mediated macrophage inflammasome activation, and also suggested EGCG administration as a promising therapeutic strategy for this disease.
Collapse
Affiliation(s)
- Minjing He
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Tianhao Chu
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ziteng Wang
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Ying Feng
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Runhan Shi
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Muyang He
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Siheng Feng
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Lin Lu
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Chen Cai
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Fang Fang
- Department of Dermatology, Shanghai Eighth People’s Hospital, Shanghai, China
| | - Xuemin Zhang
- Department of Trauma Emergency & Critical Care Medicine, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
- *Correspondence: Bo Gao, ; Yi Liu, ; Xuemin Zhang,
| | - Yi Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Bo Gao, ; Yi Liu, ; Xuemin Zhang,
| | - Bo Gao
- Department of Immunology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
- *Correspondence: Bo Gao, ; Yi Liu, ; Xuemin Zhang,
| |
Collapse
|
7
|
Machin A, Susilo I, Purwanto DA. Green tea and its active compound epigallocathechin-3-gallate (EGCG) inhibit neuronal apoptosis in a middle cerebral artery occlusion (MCAO) model. J Basic Clin Physiol Pharmacol 2021; 32:319-325. [PMID: 34214383 DOI: 10.1515/jbcpp-2020-0454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 02/20/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To determine the effect of green tea with the active ingredient epigallocathechin-3-gallate (EGCG) on the inhibition of apoptosis in the middle cerebral artery occlusion (MCAO) model. METHODS Four month old male Rattus norvegicus rats with a body weight of 200-275 g was used for the MCAO model and divided into five groups, and the treatment was carried out for 7 days. Before being sacrificed, the subject had 1 cc of blood drawn for high mobility group box 1 (HMGB-1) examination using enzyme-linked immunosorbent assay (ELISA), and after being sacrificed, the brain tissue specimen was taken to examine caspase-3 and B-cell lymphoma 3 (BCL-3) using immunohistochemistry methods. RESULTS There was no significant difference in HMGB-1 results for the treatment group compared to the control group (P1: 384.20 ± 231.72 [p = 0.553]; P2: 379.11 ± 268.4 [p = 0.526]; P3: 284, 87 ± 276.19 [p = 0.140]; P4: 435.32 ± 279.95 [p = 0.912]). There is a significant increase in BCL-2 expression between the treatment group compared to the control group (P1: 2.58 ± 0.51 [p = 0.04]; P2: 3.36 ± 0.50 [p<0.001]; P3: 4.00 ± 0.42 [p<0.001]; P4: 3.60 ± 0.52 [p<0.001]). There was a significant difference in caspase-3 expression compared to the control group in the P3 group (P1: 4.33 ± 0.49 [p = 0.652]; P2: 4.09 ± 0.30 [p = 0.136]; P3: 3.58 ± 0.51 [p = 0.01]; P4: 3.89 ± 0.42 [p = 0.063]). There is no correlation between HMGB-1 and caspase-3 (r = -0.063; p = 0.613) or BCL-2 (r = -0.106; p = 0.396). There is significant negative correlation between caspase-3 and BCL-2 (r = -0.459; p = 0.000). CONCLUSIONS Green tea with the active ingredient EGCG can inhibit neuronal cell death through the apoptotic pathway and not through the activation of HMGB-1.
Collapse
Affiliation(s)
- Abdulloh Machin
- Department Neurology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Imam Susilo
- Department Clinical Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Djoko A Purwanto
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
8
|
Wang X, Greenblatt HM, Bigman LS, Yu B, Pletka CC, Levy Y, Iwahara J. Dynamic Autoinhibition of the HMGB1 Protein via Electrostatic Fuzzy Interactions of Intrinsically Disordered Regions. J Mol Biol 2021; 433:167122. [PMID: 34181980 DOI: 10.1016/j.jmb.2021.167122] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/03/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
Highly negatively charged segments containing only aspartate or glutamate residues ("D/E repeats") are found in many eukaryotic proteins. For example, the C-terminal 30 residues of the HMGB1 protein are entirely D/E repeats. Using nuclear magnetic resonance (NMR), fluorescence, and computational approaches, we investigated how the D/E repeats causes the autoinhibition of HMGB1 against its specific binding to cisplatin-modified DNA. By varying ionic strength in a wide range (40-900 mM), we were able to shift the conformational equilibrium between the autoinhibited and uninhibited states toward either of them to the full extent. This allowed us to determine the macroscopic and microscopic equilibrium constants for the HMGB1 autoinhibition at various ionic strengths. At a macroscopic level, a model involving the autoinhibited and uninhibited states can explain the salt concentration-dependent binding affinity data. Our data at a microscopic level show that the D/E repeats and other parts of HMGB1 undergo electrostatic fuzzy interactions, each of which is weaker than expected from the macroscopic autoinhibitory effect. This discrepancy suggests that the multivalent nature of the fuzzy interactions enables strong autoinhibition at a macroscopic level despite the relatively weak intramolecular interaction at each site. Both experimental and computational data suggest that the D/E repeats interact preferentially with other intrinsically disordered regions (IDRs) of HMGB1. We also found that mutations mimicking post-translational modifications relevant to nuclear export of HMGB1 can moderately modulate DNA-binding affinity, possibly by impacting the autoinhibition. This study illuminates a functional role of the fuzzy interactions of D/E repeats.
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, TX 77555-1068, USA
| | - Harry M Greenblatt
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Lavi S Bigman
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Binhan Yu
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555-1068, USA
| | - Channing C Pletka
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555-1068, USA
| | - Yaakov Levy
- Department of Chemical and Structural Biology, Weizmann Institute of Science, Rehovot 76100, Israel.
| | - Junji Iwahara
- Department of Biochemistry and Molecular Biology, Sealy Center for Structural Biology and Molecular Biophysics, University of Texas Medical Branch, Galveston, TX 77555-1068, USA.
| |
Collapse
|
9
|
Sekiguchi F, Kawabata A. Role of HMGB1 in Chemotherapy-Induced Peripheral Neuropathy. Int J Mol Sci 2020; 22:ijms22010367. [PMID: 33396481 PMCID: PMC7796379 DOI: 10.3390/ijms22010367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/25/2020] [Accepted: 12/26/2020] [Indexed: 12/11/2022] Open
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN), one of major dose-limiting side effects of first-line chemotherapeutic agents such as paclitaxel, oxaliplatin, vincristine, and bortezomib is resistant to most of existing medicines. The molecular mechanisms of CIPN have not been fully understood. High mobility group box 1 (HMGB1), a nuclear protein, is a damage-associated molecular pattern protein now considered to function as a pro-nociceptive mediator once released to the extracellular space. Most interestingly, HMGB1 plays a key role in the development of CIPN. Soluble thrombomodulin (TMα), known to degrade HMGB1 in a thrombin-dependent manner, prevents CIPN in rodents treated with paclitaxel, oxaliplatin, or vincristine and in patients with colorectal cancer undergoing oxaliplatin-based chemotherapy. In this review, we describe the role of HMGB1 and its upstream/downstream mechanisms in the development of CIPN and show drug candidates that inhibit the HMGB1 pathway, possibly useful for prevention of CIPN.
Collapse
|
10
|
Kwak MS, Kim HS, Lee B, Kim YH, Son M, Shin JS. Immunological Significance of HMGB1 Post-Translational Modification and Redox Biology. Front Immunol 2020; 11:1189. [PMID: 32587593 PMCID: PMC7297982 DOI: 10.3389/fimmu.2020.01189] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/13/2020] [Indexed: 12/19/2022] Open
Abstract
Most extracellular proteins are secreted via the classical endoplasmic reticulum (ER)/Golgi-dependent secretion pathway; however, some proteins, including a few danger-associated molecular patterns (DAMPs), are secreted via non-classical ER/Golgi-independent secretion pathways. The evolutionarily conserved high mobility group box1 (HMGB1) is a ubiquitous nuclear protein that can be released by almost all cell types. HMGB1 lacks signal peptide and utilizes diverse non-canonical secretion mechanisms for its extracellular export. Although the post-translational modifications of HMGB1 were demonstrated, the oxidation of HMGB1 and secretion mechanisms are not highlighted yet. We currently investigated that peroxiredoxins I and II (PrxI/II) induce the intramolecular disulfide bond formation of HMGB1 in the nucleus. Disulfide HMGB1 is preferentially transported out of the nucleus by binding to the nuclear exportin chromosome-region maintenance 1 (CRM1). We determined the kinetics of HMGB1 oxidation in bone marrow-derived macrophage as early as a few minutes after lipopolysaccharide treatment, peaking at 4 h while disulfide HMGB1 accumulation was observed within the cells, starting to secrete in the late time point. We have shown that HMGB1 oxidation status, which is known to determine the biological activity in extracellular HMGB1, is crucial for the secretion of HMGB1 from the nucleus. This review summarizes selected aspects of HMGB1 redox biology relevant to the induction and propagation of inflammatory diseases. We implicate the immunological significance and the need for novel HMGB1 inhibitors through mechanism-based studies.
Collapse
Affiliation(s)
- Man Sup Kwak
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Hee Sue Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Bin Lee
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young Hun Kim
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea
| | - Myoungsun Son
- Center for Autoimmune Musculoskeletal and Hematopoietic Diseases, Institute of Molecular Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, United States
| | - Jeon-Soo Shin
- Department of Microbiology, Yonsei University College of Medicine, Seoul, South Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.,Center for Nanomedicine, Institute for Basic Science (IBS), Yonsei University, Seoul, South Korea
| |
Collapse
|
11
|
Tsujita R, Tsubota M, Sekiguchi F, Kawabata A. Role of high-mobility group box 1 and its modulation by thrombomodulin/thrombin axis in neuropathic and inflammatory pain. Br J Pharmacol 2020; 178:798-812. [PMID: 32374414 DOI: 10.1111/bph.15091] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022] Open
Abstract
High-mobility group box 1 (HMGB1), a nuclear protein, once released to the extracellular space, facilitates pain signals as well as inflammation. Intraplantar or intraspinal application of HMGB1 elicits hyperalgesia/allodynia in rodents by activating the advanced glycosylation end-product specific receptor (receptor for advanced glycation end-products; RAGE) or Toll-like receptor 4 (TLR4). Endogenous HMGB1 derived from neurons, perineuronal cells or immune cells accumulating in the dorsal root ganglion or sensory nerves participates in somatic and visceral pain consisting of neuropathic and/or inflammatory components. Endothelial thrombomodulin (TM) and recombinant human soluble TM, TMα, markedly increase thrombin-dependent degradation of HMGB1, and systemic administration of TMα prevents and reverses various HMGB1-dependent pathological pain. Low MW compounds that directly inactivate HMGB1 or antagonize HMGB1-targeted receptors would be useful to reduce various forms of intractable pain. Thus, HMGB1 and its receptors are considered to serve as promising targets in developing novel agents to prevent or treat pathological pain. LINKED ARTICLES: This article is part of a themed issue on Neurochemistry in Japan. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.4/issuetoc.
Collapse
Affiliation(s)
- Ryuichi Tsujita
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formally known as Kinki University), Higashiosaka, Japan.,Project Management Department, Asahi Kasei Pharma Corporation, Tokyo, Japan
| | - Maho Tsubota
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formally known as Kinki University), Higashiosaka, Japan
| | - Fumiko Sekiguchi
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formally known as Kinki University), Higashiosaka, Japan
| | - Atsufumi Kawabata
- Laboratory of Pharmacology and Pathophysiology, Faculty of Pharmacy, Kindai University (formally known as Kinki University), Higashiosaka, Japan
| |
Collapse
|
12
|
Anggayasti WL, Ogino K, Yamamoto E, Helmerhorst E, Yasuoka K, Mancera RL. The acidic tail of HMGB1 regulates its secondary structure and conformational flexibility: A circular dichroism and molecular dynamics simulation study. Comput Struct Biotechnol J 2020; 18:1160-1172. [PMID: 32514327 PMCID: PMC7261955 DOI: 10.1016/j.csbj.2020.05.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/06/2020] [Accepted: 05/09/2020] [Indexed: 12/02/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a damage-associated molecular pattern (DAMP) molecule that triggers the progression of several pro-inflammatory diseases such as diabetes, Alzheimer's disease and cancer, by inducing signals upon interaction with the receptors such as the receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs). The acidic C-terminal tail of HMGB1 is an intrinsically disordered region of the protein which is known to determine the interaction of HMGB1 to DNA and histones. This study characterizes its structural properties using a combination of circular dichroism (CD) and molecular dynamics (MD) simulations. The full-length and tail-less forms of HMGB1 were compared to rationalise the role of the acidic tail in maintaining the stability of the entire structure of HMGB1 in atomistic detail. Consistent with experimental data, the acidic tail was predicted to adopt an extended conformation that allows it to make a range of hydrogen-bonding and electrostatic interactions with the box-like domains that stabilize the overall structure of HMGB1. Absence of the acidic tail was predicted to increase structural fluctuations of all amino acids, leading to changes in secondary structure from α-helical to more hydrophilic turns along with increased exposure of multiple amino acids to the surrounding solvent. These structural changes reveal the intrinsic conformational dynamics of HMGB1 that are likely to affect the accessibility of its receptors.
Collapse
Affiliation(s)
- Wresti L. Anggayasti
- Department of Chemical Engineering, Faculty of Engineering, Brawijaya University, Jl. MT Haryono 167, Malang 65145, East Java, Indonesia
| | - Kenta Ogino
- Department of Mechanical Engineering, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Eiji Yamamoto
- Department of System Design Engineering, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Erik Helmerhorst
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Kenji Yasuoka
- Department of Mechanical Engineering, Keio University, Yokohama, Kanagawa 223-8522, Japan
| | - Ricardo L. Mancera
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute and Curtin Institute for Computation, Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| |
Collapse
|
13
|
Nugraha AP, Narmada IB, Sitasari PI, Inayati F, Wira R, Triwardhani A, Hamid T, Ardani IGAW, Djaharu’ddin I, Rahmawati D, Iskandar RPD. High Mobility Group Box 1 and Heat Shock Protein-70 Expression Post (-)-Epigallocatechin-3-Gallate in East Java Green Tea Methanolic Extract Administration During Orthodontic Tooth Movement in Wistar Rats. PESQUISA BRASILEIRA EM ODONTOPEDIATRIA E CLÍNICA INTEGRADA 2020. [DOI: 10.1590/pboci.2020.040] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
14
|
Chu B, Wu S, Ji X, Chen R, Song B, Tang J, Wang H, Su Y, He Y. Controllable silicon nanostructures featuring stable fluorescence and intrinsic in vitro and in vivo anti-cancer activity. J Mater Chem B 2019; 7:6247-6256. [PMID: 31566627 DOI: 10.1039/c9tb01191a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
In this manuscript, we demonstrate that the in situ growth of fluorescent silicon (Si) nanomaterials is stimulated when organosilicane molecules interact with different green teas, producing multifunctional Si nanomaterials with controllable zero- (e.g., nanoparticles), two- (e.g., nanosheets), and three- (e.g., nanospheres) dimensional nanostructures. Such green tea-originated Si nanomaterials (GTSN) exhibit strong fluorescence (quantum yield: ∼19-30%) coupled with ultrahigh photostability, as well as intrinsic anti-cancer activity with high specificity (e.g., the GTSN can accurately kill various cancer cells, rather than normal cells). Taking advantage of these unique merits, we further performed systematic in vitro and in vivo experiments to interrogate the mechanism of the green tea- and GTSN-related cancer prevention. Typically, we found that the GTSN entered the cell nuclei and induced cell apoptosis/death of cancer cells. The prepared GTSN were observed in vivo to accumulate in the tumour tissues after 14-d post-injection, leading to an efficient inhibition of tumour growth. Our results open new avenues for designing novel multifunctional and side-effect-free Si nanomaterials with controllable structures.
Collapse
Affiliation(s)
- Binbin Chu
- Laboratory of Nanoscale Biochemical Analysis, Institute of Functional Nano & Soft Materials (FUNSOM), and Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, Jiangsu 215123, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Wang Z, Han Q, Guo YL, Liu XH, Qiu T. Effect of ozone oxidative preconditioning on inflammation and oxidative stress injury in rat model of renal transplantation. Acta Cir Bras 2018; 33:238-249. [PMID: 29668774 DOI: 10.1590/s0102-865020180030000006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/22/2018] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To investigate the effect of ozone oxidative preconditioning (OzoneOP) on inflammation and oxidative stress injury in rat model of renal transplantation. METHODS Thirty six male Sprague Dawley (SD) rats were randomly divided into three groups. Sham group: rats were treated with opening and closing abdomen. Kidney transplantation group (KT group): SD rat received the donor's left kidney derived from another SD rat. Ozone oxidative preconditioning and kidney transplantation (OOP+KT group): donor SD rats received OzoneOP treatments by transrectal insufflations before kidney transplantation. After transplantation, parameters of renal function of recipients were determined. Morphology and pathological changes of renal allograft were examined. Expression of NF-κBp65, HMGB-1 were also determined by Western-blot. RESULTS Compared to KT group, the morphology and pathological damages of renal allograft were less serious in OOP+KT group. Meanwhile, levels of SOD and GSH-Px of renal allograft in OOP+KT group were higher than those in KT group respectively. Western-blot showed that the expressions of NF-κBp65 and HMGB-1 in OOP+KT group were obviously less than those in KT group. CONCLUSION Ozone oxidative preconditioning could attenuate the inflammatory reaction and oxidative stress injury in renal allograft, which might be related with the enhancement of anti-oxidative system and suppression of inflammatory reaction.
Collapse
Affiliation(s)
- Zhishun Wang
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Han
- Department of Nephrology, The Fifth Hospital of Wuhan, Wuhan, China
| | - Yong-Lian Guo
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiu-Heng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
16
|
VanPatten S, Al-Abed Y. High Mobility Group Box-1 (HMGb1): Current Wisdom and Advancement as a Potential Drug Target. J Med Chem 2018; 61:5093-5107. [PMID: 29268019 DOI: 10.1021/acs.jmedchem.7b01136] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
High mobility group box-1 (HMGb1) protein, a nuclear non-histone protein that is released or secreted from the cell in response to damage or stress, is a sentinel for the immune system that plays a critical role in cell survival/death pathways. This review highlights key features of the endogenous danger-associated molecular pattern (DAMP) protein, HMGb1 in the innate inflammatory response along with various cofactors and receptors that regulate its downstream effects. The evidence demonstrating increased levels of HMGb1 in human inflammatory diseases and conditions is presented, along with a summary of current small molecule or peptide-like antagonists proven to specifically target HMGb1. Additionally, we delineate the measures needed toward validating this protein as a clinically relevant biomarker or bioindicator and as a relevant drug target.
Collapse
Affiliation(s)
- Sonya VanPatten
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| | - Yousef Al-Abed
- Center for Molecular Innovation , The Feinstein Institute for Medical Research , 350 Community Drive , Manhasset , New York 11030 , United States
| |
Collapse
|
17
|
Zhao L, Liu S, Xu J, Li W, Duan G, Wang H, Yang H, Yang Z, Zhou R. A new molecular mechanism underlying the EGCG-mediated autophagic modulation of AFP in HepG2 cells. Cell Death Dis 2017; 8:e3160. [PMID: 29095434 PMCID: PMC5775413 DOI: 10.1038/cddis.2017.563] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/20/2017] [Accepted: 09/22/2017] [Indexed: 01/26/2023]
Abstract
Epigallocatechingallate (EGCG) is a major bioactive component of green tea and is associated with health benefits against multiple diseases including cancer. As an indicator of hepatocellular carcinoma (HCC), high levels of α-fetal protein (AFP) are related to malignant differentiation and poor prognosis of cancer cells. In this study, EGCG can effectively reduce AFP secretion and simultaneously induce AFP aggregation in human HCC HepG2 cells. EGCG-stimulated autophagy induces the degradation of AFP aggregates in HepG2 cells. Furthermore, we thoroughly studied the underlying molecular mechanisms behind EGCG-stimulated autophagy by using large-scale all-atom molecular dynamics simulations, which revealed a novel molecular mechanism. EGCG directly interacts with LC3-I protein, readily exposing the pivotal Gly-120 site of the latter to other important binding partners such as 1,2-distearoyl-sn-glycero-3-phosphoethanolamine and promoting the synthesis of LC3-II, a characteristic autophagosomal marker. Our results suggest that EGCG is critical in regulating AFP secretion and in modulating autophagic activities of HepG2 cells, providing a molecular basis for potentially preventing and treating HCC.
Collapse
Affiliation(s)
- Lin Zhao
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Shengtang Liu
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiaying Xu
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Wei Li
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - Guangxin Duan
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Haichao Wang
- The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Zaixing Yang
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ruhong Zhou
- School of Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.,Computational Biological Center, IBM Thomas J Watson Research Center, Yorktown Heights, NY 10598, USA.,Department of Chemistry, Columbia University, New York, NY 10027, USA
| |
Collapse
|
18
|
Anggayasti WL, Mancera RL, Bottomley S, Helmerhorst E. The self-association of HMGB1 and its possible role in the binding to DNA and cell membrane receptors. FEBS Lett 2017; 591:282-294. [PMID: 28027393 DOI: 10.1002/1873-3468.12545] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/04/2016] [Accepted: 12/22/2016] [Indexed: 01/09/2023]
Abstract
High mobility group box 1 (HMGB1), a chromatin protein, interacts with DNA and controls gene expression. However, when HMGB1 is released from apoptotic or damaged cells, it triggers proinflammatory reactions by interacting with various receptors, mainly receptor for advanced glycation end-products (RAGE) and toll-like receptors (TLRs). The self-association of HMGB1 has been found to be crucial for its DNA-related biological functions. It is influenced by several factors, such as ionic strength, pH, specific divalent metal cations, redox environment and acetylation. This self-association may also play a role in the interaction with RAGE and TLRs and the concomitant inflammatory responses. Future studies should address the potential role of HMGB1 self-association on its interactions with DNA, RAGE and TLRs, as well as the influence of physicochemical factors in different cellular environments on these interactions.
Collapse
Affiliation(s)
- Wresti L Anggayasti
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Ricardo L Mancera
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Steve Bottomley
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| | - Erik Helmerhorst
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, Australia
| |
Collapse
|
19
|
Panneerselvam S, Durai P, Yesudhas D, Achek A, Kwon HK, Choi S. Cysteine redox state plays a key role in the inter-domain movements of HMGB1: a molecular dynamics simulation study. RSC Adv 2016. [DOI: 10.1039/c6ra16343b] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We have modelled and simulated different states of HMGB1, suggesting that the fully reduced HMGB1 maintains the inter-domain movements during the activity.
Collapse
Affiliation(s)
| | | | - Dhanusha Yesudhas
- Department of Molecular Science and Technology
- Ajou University
- Suwon 443-749
- Korea
| | - Asma Achek
- Department of Molecular Science and Technology
- Ajou University
- Suwon 443-749
- Korea
| | - Hyuk-Kwon Kwon
- Department of Molecular Science and Technology
- Ajou University
- Suwon 443-749
- Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology
- Ajou University
- Suwon 443-749
- Korea
| |
Collapse
|