1
|
Pluma-Pluma A, García G, Murbartián J. Chronic restraint stress and social transfer of stress produce tactile allodynia mediated by the HMGB1/TNFα/TNFR1 pathway in female and male rats. Physiol Behav 2024; 274:114418. [PMID: 38042454 DOI: 10.1016/j.physbeh.2023.114418] [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: 09/13/2023] [Revised: 10/17/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Previous studies have shown the relevance of high mobility group box 1 protein (HMGB1) and tumor necrosis factor α (TNFα) in nerve or tissue injury-induced nociception. However, the role of these proteins in chronic stress and social transfer of stress (STS)-induced dysfunctional pain is not entirely known. The aim of this study was to determine the participation of the spinal HMGB1-TNFα signaling pathway and TNFα receptor 1 (TNFR1) in rats subjected to chronic restraint stress (CRS) and STS. Non-stressed female and male rats in contact with CRS rats increased sniffing behavior of the anogenital area, behavior related to STS. Rats subjected to CRS and STS reduced 50 % withdrawal threshold and reached the value of tactile allodynia after 21 days of stress. Rats return to the basal withdrawal threshold after 30 days without stress and return to allodynia values in only 5 days of stress sessions (priming). Female and male rats subjected to 28 days of CRS or STS were intrathecal injected with glycyrrhizin (inhibitor of HMGB1), thalidomide (inhibitor of the TNFα synthesis), and R7050 (TNFR1 antagonist), in all the cases, an antiallodynic effect was observed. Rats under CRS or STS enhanced HMGB1 and TNFR1 protein expression in DRG and dorsal spinal cord. Data suggest that the spinal HMGB1/TNFα/TNFR1 signaling pathway plays a relevant role in the maintenance of CRS and STS-induced nociceptive hypersensitivity in rats. These proteins could be helpful in developing pain treatments for fibromyalgia in humans.
Collapse
Affiliation(s)
- Alejandro Pluma-Pluma
- Departamento de Farmacobiología, Cinvestav, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, South Campus, Mexico City, Mexico
| | - Guadalupe García
- Departamento de Farmacobiología, Cinvestav, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, South Campus, Mexico City, Mexico
| | - Janet Murbartián
- Departamento de Farmacobiología, Cinvestav, Calzada de los Tenorios 235, Colonia Granjas Coapa, 14330, South Campus, Mexico City, Mexico.
| |
Collapse
|
2
|
Yang JP, Ullah A, Su YN, Otoo A, Adu-Gyamfi EA, Feng Q, Wang YX, Wang MJ, Ding YB. Glycyrrhizin ameliorates impaired glucose metabolism and ovarian dysfunction in a polycystic ovary syndrome mouse model. Biol Reprod 2023; 109:83-96. [PMID: 37115805 DOI: 10.1093/biolre/ioad048] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 03/24/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023] Open
Abstract
The aim of this study was to determine the impact of glycyrrhizin, an inhibitor of high mobility group box 1, on glucose metabolic disorders and ovarian dysfunction in mice with polycystic ovary syndrome. We generated a polycystic ovary syndrome mouse model by using dehydroepiandrosterone plus high-fat diet. Glycyrrhizin (100 mg/kg) was intraperitoneally injected into the polycystic ovary syndrome mice and the effects on body weight, glucose tolerance, insulin sensitivity, estrous cycle, hormone profiles, ovarian pathology, glucolipid metabolism, and some molecular mechanisms were investigated. Increased number of cystic follicles, hormonal disorders, impaired glucose tolerance, and decreased insulin sensitivity in the polycystic ovary syndrome mice were reverted by glycyrrhizin. The increased high mobility group box 1 levels in the serum and ovarian tissues of the polycystic ovary syndrome mice were also reduced by glycyrrhizin. Furthermore, increased expressions of toll-like receptor 9, myeloid differentiation factor 88, and nuclear factor kappa B as well as reduced expressions of insulin receptor, phosphorylated protein kinase B, and glucose transporter type 4 were restored by glycyrrhizin in the polycystic ovary syndrome mice. Glycyrrhizin could suppress the polycystic ovary syndrome-induced upregulation of high mobility group box 1, several inflammatory marker genes, and the toll-like receptor 9/myeloid differentiation factor 88/nuclear factor kappa B pathways, while inhibiting the insulin receptor/phosphorylated protein kinase B/glucose transporter type 4 pathways. Hence, glycyrrhizin is a promising therapeutic agent against polycystic ovary syndrome.
Collapse
Affiliation(s)
- Jun-Pu Yang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Amin Ullah
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Ya-Nan Su
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Antonia Otoo
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | - Qian Feng
- Department of Obstetrics and Gynecology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Ying-Xiong Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Mei-Jiao Wang
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Physiology, School of Basic Medical Science, Chongqing Medical University, Chongqing, China
| | - Yu-Bin Ding
- Department of Obstetrics and Gynecology, Women and Children's Hospital of Chongqing Medical University, Chongqing, China
- Department of Toxicology, School of Public Health, Chongqing Medical University, Chongqing, China
- Department of Pharmacology, Academician Workstation, Changsha Medical University, Changsha, China
| |
Collapse
|
3
|
Singh KP, Pallett LJ, Singh H, Chen A, Otano I, Duriez M, Rombouts K, Pinzani M, Crane M, Fusai G, Avihingsanon A, Lewin SR, Maini MK. Pro-fibrogenic role of alarmin high mobility group box 1 in HIV-hepatitis B virus coinfection. AIDS 2023; 37:401-411. [PMID: 36384811 DOI: 10.1097/qad.0000000000003435] [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: 11/18/2022]
Abstract
OBJECTIVE Liver disease is accelerated in people with HIV (PWH) with hepatitis B virus (HBV) coinfection. We hypothesized that liver fibrosis in HIV-HBV is triggered by increased hepatocyte apoptosis, microbial translocation and/or HIV/HBV viral products. DESIGN Sera from PWH with HBV coinfection versus from those with HBV only or putative mediators were used to examine the pathogenesis of liver disease in HIV-HBV. METHODS We applied sera from PWH and HBV coinfection versus HBV alone, or putative mediators (including HMGB1), to primary human hepatic stellate cells (hHSC) and examined pro-fibrogenic changes at the single cell level using flow cytometry. High mobility group box 1 (HMGB1) levels in the applied sera were assessed according to donor fibrosis stage. RESULTS Quantitative flow cytometric assessment of pro-fibrogenic and inflammatory changes at the single cell level revealed an enhanced capacity for sera from PWH with HBV coinfection to activate hHSC. This effect was recapitulated by lipopolysaccharide, HIV-gp120, hepatocyte conditioned-media and the alarmin HMGB1. Induction of hepatocyte cell death increased their pro-fibrogenic potential, an effect blocked by HMGB1 antagonist glycyrrhizic acid. Consistent with a role for this alarmin, HMGB1 levels were elevated in sera from PWH and hepatitis B coinfection compared to HBV alone and higher in those with HIV-HBV with liver fibrosis compared to those without. CONCLUSIONS Sera from PWH and HBV coinfection have an enhanced capacity to activate primary hHSC. We identified an increase in circulating HMGB1 which, in addition to HIV-gp120 and translocated microbial products, drove pro-fibrogenic changes in hHSC, as mechanisms contributing to accelerated liver disease in HIV-HBV.
Collapse
Affiliation(s)
- Kasha P Singh
- Division of Infection and Immunity, University College London, London, UK
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Laura J Pallett
- Division of Infection and Immunity, University College London, London, UK
| | - Harsimran Singh
- Division of Infection and Immunity, University College London, London, UK
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Antony Chen
- Division of Infection and Immunity, University College London, London, UK
| | - Itziar Otano
- Division of Infection and Immunity, University College London, London, UK
| | - Marion Duriez
- Division of Infection and Immunity, University College London, London, UK
| | - Krista Rombouts
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Massimo Pinzani
- Institute for Liver and Digestive Health, University College London, London, UK
| | - Megan Crane
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
| | - Giuseppe Fusai
- Institute for Liver and Digestive Health, University College London, London, UK
| | | | - Sharon R Lewin
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity
- Department of Infectious Diseases, Alfred Health and Monash University, Melbourne, Victoria, Australia
| | - Mala K Maini
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
4
|
Di Bartolomeo L, Custurone P, Irrera N, Borgia F, Vaccaro F, Squadrito F, Vaccaro M. Vitiligo and Mental Health: Natural Compounds' Usefulness. Antioxidants (Basel) 2023; 12:antiox12010176. [PMID: 36671038 PMCID: PMC9854903 DOI: 10.3390/antiox12010176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Vitiligo is an autoimmune dermatosis frequently associated with other comorbidities, such as mental health disorders. It is unclear if vitiligo triggers mental disorders or if mental disorders trigger vitiligo, but each one affects and worsen the other, if present at the same time. Both mental health disorders and vitiligo present a multifactorial pathogenesis and often require prolonged periods of therapy, sometimes with poor results. Given the possible link of common pathogenetic factors and the need of integrated therapies, the aim of this review is to look at natural compounds as possible supplements for both conditions. The results yielded show a possible role of these supplements in ameliorating both conditions, thus helping these patients to achieve a better quality of life and reduce the need for prolonged therapies. The limitations regarding the relative lack of in vivo studies, and the increasing need to lighten the burden of these chronic diseases, suggests that it is mandatory to proceed with further trials.
Collapse
Affiliation(s)
- Luca Di Bartolomeo
- Department of Clinical and Experimental Medicine, Dermatology, University of Messina, 98125 Messina, Italy
| | - Paolo Custurone
- Department of Clinical and Experimental Medicine, Dermatology, University of Messina, 98125 Messina, Italy
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, Pharmacology, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Francesco Borgia
- Department of Clinical and Experimental Medicine, Dermatology, University of Messina, 98125 Messina, Italy
| | - Federico Vaccaro
- Department of Dermatology, University of Modena and Reggio Emilia, 41121 Modena, Italy
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, Pharmacology, University of Messina, 98125 Messina, Italy
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, Dermatology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
5
|
Chen B, Qin G, Xiao J, Deng X, Lin A, Liu H. Transient neuroinflammation following surgery contributes to long-lasting cognitive decline in elderly rats via dysfunction of synaptic NMDA receptor. J Neuroinflammation 2022; 19:181. [PMID: 35831873 PMCID: PMC9281167 DOI: 10.1186/s12974-022-02528-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 06/12/2022] [Indexed: 11/27/2022] Open
Abstract
Background Perioperative neurocognitive disorders (PNDs) are considered the most common postoperative complication in geriatric patients. However, its pathogenesis is not fully understood. Surgery-triggered neuroinflammation is a major contributor to the development of PNDs. Neuroinflammation can influence N-methyl-D-aspartate receptor (NMDAR) expression or function which is closely associated with cognition. We, therefore, hypothesized that the persistent changes in NMDAR expression or function induced by transient neuroinflammation after surgery were involved in the development of PNDs. Methods Eighteen-month-old male Sprague–Dawley rats were subjected to abdominal surgery with sevoflurane anesthesia to establish the PNDs animal model. Then, we determined the transient neuroinflammation by detecting the protein levels of proinflammatory cytokines and microglia activation using ELISA, western blot, immunohistochemistry, and microglial morphological analysis from postoperative days 1–20. Persistent changes in NMDAR expression were determined by detecting the protein levels of NMDAR subunits from postoperative days 1–59. Subsequently, the dysfunction of synaptic NMDAR was evaluated by detecting the structural plasticity of dendritic spine using Golgi staining. Pull-down assay and western blot were used to detect the protein levels of Rac1-GTP, phosphor-cofilin, and Arp3, which contribute to the regulation of the structural plasticity of dendritic spine. Finally, glycyrrhizin, an anti-inflammatory agent, was administered to further explore the role of synaptic NMDAR dysfunction induced by transient neuroinflammation in the neuropathogenesis of PNDs. Results We showed that transient neuroinflammation induced by surgery caused sustained downregulation of synaptic NR2A and NR2B subunits in the dorsal hippocampus and led to a selective long-term spatial memory deficit. Meanwhile, the detrimental effect of neuroinflammation on the function of synaptic NMDARs was shown by the impaired structural plasticity of dendritic spines and decreased activity of the Rac1 signaling pathways during learning. Furthermore, anti-inflammatory treatment reversed the downregulation and hypofunction of synaptic NR2A and NR2B and subsequently rescued the long-term spatial memory deficit. Conclusions Our results identify sustained synaptic NR2A and NR2B downregulation and hypofunction induced by transient neuroinflammation following surgery as important contributors to the development of PNDs in elderly rats. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02528-5.
Collapse
Affiliation(s)
- Bo Chen
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Guangcheng Qin
- Laboratory Research Center, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jingyu Xiao
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Xiaoyuan Deng
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China
| | - Aolei Lin
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Hongliang Liu
- Department of Anesthesiology, Chongqing University Cancer Hospital, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
6
|
Hisaoka-Nakashima K, Ohata K, Yoshimoto N, Tokuda S, Yoshii N, Nakamura Y, Wang D, Liu K, Wake H, Yoshida T, Ago Y, Hashimoto K, Nishibori M, Morioka N. High-mobility group box 1-mediated hippocampal microglial activation induces cognitive impairment in mice with neuropathic pain. Exp Neurol 2022; 355:114146. [PMID: 35738416 DOI: 10.1016/j.expneurol.2022.114146] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 11/28/2022]
Abstract
Clinical evidence indicates that cognitive impairment is a common comorbidity of chronic pain, including neuropathic pain, but the mechanism underlying cognitive impairment remains unclear. Neuroinflammation plays a critical role in the development of both neuropathic pain and cognitive impairment. High-mobility group box 1 (HMGB1) is a proinflammatory molecule and could be involved in neuroinflammation-mediated cognitive impairment in the neuropathic pain state. Hippocampal microglial activation in mice has been associated with cognitive impairment. Thus, the current study examined a potential role of HMGB1 and microglial activation in cognitive impairment in mice with neuropathic pain due to a partial sciatic nerve ligation (PSNL). Mice developed cognitive impairment over two weeks, but not one week, after nerve injury. Nerve-injured mice demonstrated decreased nuclear fraction HMGB1, suggesting increased extracellular release of HMGB1. Furthermore, two weeks after PSNL, significant microglia activation was observed in hippocampus. Inhibition of microglial activation with minocycline, local hippocampal microglia depletion with clodronate liposome, or blockade of HMGB1 with either glycyrrhizic acid (GZA) or anti-HMGB1 antibody in PSNL mice reduced hippocampal microglia activation and ameliorated cognitive impairment. Other changes in the hippocampus of PSNL mice potentially related to cognitive impairment, including decreased hippocampal neuron dendrite length and spine densities and decreased α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptor (AMPAR) subunits, were prevented with anti-HMGB1 antibody treatment. The current findings suggest that neuro-inflammation involves a number of cellular-level changes and microglial activation. Blocking neuro-inflammation, particularly through blocking HMGB1 could be a novel approach to reducing co-morbidities such as cognitive impairment associated with neuropathic pain.
Collapse
Affiliation(s)
- Kazue Hisaoka-Nakashima
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Kazuto Ohata
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Natsuki Yoshimoto
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Shintarou Tokuda
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Nanako Yoshii
- Department of Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical and 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
| | - Hidenori Wake
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Takayuki Yoshida
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Yukio Ago
- Department of Cellular and Molecular Pharmacology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Kouichi Hashimoto
- Department of Neurophysiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, 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 and Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
7
|
Sun S, Cai B, Li Y, Su W, Zhao X, Gong B, Li Z, Zhang X, Wu Y, Chen C, Tsang SH, Yang J, Li X. HMGB1 and Caveolin-1 related to RPE cell senescence in age-related macular degeneration. Aging (Albany NY) 2020; 11:4323-4337. [PMID: 31284269 PMCID: PMC6660032 DOI: 10.18632/aging.102039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 06/20/2019] [Indexed: 01/09/2023]
Abstract
Accumulation of lipofuscin in the retinal pigment epithelium (RPE) is considered a major cause of RPE dysfunction and senescence in age-related macular degeneration (AMD), and N-retinylidene-N-retinylethanolamine (A2E) is the main fluorophore identified in lipofuscin from aged human eyes. Here, human-induced pluripotent stem cell (iPSC)-RPE was generated from healthy individuals to reveal proteomic changes associated with A2E-related RPE cell senescence. A novel RPE cell senescence-related protein, high-mobility group box 1 (HMGB1), was identified based on proteomic mass spectrometry measurements on iPSC-RPE with A2E treatment. Furthermore, HMGB1 upregulated Caveolin-1, which also was related RPE cell senescence. To investigate whether changes in HMGB1 and Caveolin-1 expression under A2E exposure contribute to RPE cell senescence, human ARPE-19 cells were stimulated with A2E; expression of HMGB1, Caveolin-1, tight junction proteins and senescent phenotypes were verified. HMGB1 inhibition alleviated A2E induced cell senescence. Migration of RPE cells was evaluated. Notably, A2E less than or equal to 10μM induced both HMGB1 and Caveolin-1 protein upregulation and HMGB1 translocation, while Caveolin-1 expression was downregulated when there was more than 10μM A2E. Our data indicate that A2E-induced upregulation of HMGB1、Caveolin-1 and HMGB1 release may relate to RPE cell senescence and play a role in the pathogenesis of AMD.
Collapse
Affiliation(s)
- Shuo Sun
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Bincui Cai
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Yao Li
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.,Departments of Ophthalmology, Columbia University, New York, NY 10027, USA
| | - Wenqi Su
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xuzheng Zhao
- Tangshan Eye Hospital, Tangshan, People's Republic of China
| | - Boteng Gong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Zhiqing Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xiaomin Zhang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Yalin Wu
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, College of Medicine, Xiamen University, Xiamen City, People's Republic of China
| | - Chao Chen
- Fujian Provincial Key Laboratory of Ophthalmology and Visual Science, Eye Institute of Xiamen University, College of Medicine, Xiamen University, Xiamen City, People's Republic of China
| | - Stephen H Tsang
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY 10032, USA.,Departments of Ophthalmology, Columbia University, New York, NY 10027, USA
| | - Jin Yang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, People's Republic of China
| |
Collapse
|
8
|
Zhu Z, Guo Y, Li X, Teng S, Peng X, Zou P, Zhou S. Glycyrrhizic Acid Attenuates Balloon-Induced Vascular Injury Through Inactivation of RAGE Signaling Pathways. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2020. [DOI: 10.15212/cvia.2019.0577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Percutaneous coronary intervention is a well-established technique used to treat coronary artery disease, but the risk of coronary artery in-stent restenosis following percutaneous coronary intervention is still high. Previous studies revealed that high mobility group protein B1 (HMGB1)
plays a critical role in neointima formation. In this study, we aimed to investigate the role of glycyrrhizic acid (GA), an HMGB1 inhibitor, in the process of neointima formation and the potential mechanisms. We investigated the role of GA in neointima formation through an iliac artery balloon
injury model in rabbits. Proliferation, migration, and phenotype transformation of human vascular smooth muscle cells (VSMCs) were observed. Besides, inflammation and receptor for advanced glycosylation end products (RAGE) signaling pathways were studied. The results indicate that GA attenuated
neointima formation and downregulated HMGB1 expression in injured artery in rabbits. HMGB1 promoted proliferation, migration, and phenotype transformation through the activation of RAGE signaling pathways in VSMCs, and blockade of HMGB1 by GA (1, 10, and 100 μM) could attenuate those processes
and reduce proliferation of human VSMCs. In conclusion, the HMGB1 inhibitor GA might be useful to treat proliferative vascular diseases by downregulating RAGE signaling pathways. Our results indicate a new and promising therapeutic agent for restenosis.
Collapse
Affiliation(s)
- Zhaowei Zhu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanan Guo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xuping Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuai Teng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaofan Peng
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Pu Zou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shenghua Zhou
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Cao ZY, Liu YZ, Li JM, Ruan YM, Yan WJ, Zhong SY, Zhang T, Liu LL, Wu R, Wang B, Wang W, Bi XY, Wang YX, Su WJ, Jiang CL. Glycyrrhizic acid as an adjunctive treatment for depression through anti-inflammation: A randomized placebo-controlled clinical trial. J Affect Disord 2020; 265:247-254. [PMID: 32090748 DOI: 10.1016/j.jad.2020.01.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 12/20/2019] [Accepted: 01/12/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Recently, abundant evidence indicated proinflammatory cytokines might play a crucial role in pathophysiology and treatment of depression. According to our preclinical research, we propose glycyrrhizic acid (GZA) for an adjunctive treatment owing to its safety, economical and anti-inflammatory profile. METHODS Eligible participants were recruited and randomly allocated into independent treatment groups of SSRI+GZA (n = 30) and SSRI+PBO (placebo, n = 26). Depressive symptoms and specific serum biomarkers were detected during the 4-week treatment course. Afterward, the relationships between biomarkers and clinical effects were explored. RESULTS Depressive symptoms relieved more in SSRI+GZA than SSRI+PBO, both at week 2 (P = 0.003) and week 4 (P = 0.016). Meanwhile, at week 4, both response rate (P = 0.035) and remission rate (P = 0.031) acutely became higher in SSRI+GZA compared with SSRI+PBO. Mediation analysis further demonstrated that TNF-α reduction mediated the association between GZA treatment and clinical improvement, the indirect effect lay between 0.124 and 3.514 (95% CI). The exploratory analysis also suggested that the symptomatic improvement existed in patients with high-inflammation (baseline CRP > 3 mg/L) rather than those with low-inflammation (baseline CRP ≤ 3 mg/L). LIMITATIONS The sample size in this study was not large enough and the follow-up duration was relatively short. CONCLUSIONS This study offers a novel strategy for the diagnosis, categorization, individualization and prognosis regarding upgrading traditional antidepressant therapy, which is from biomarkers to diagnostic indicator and therapeutic target. Patients are necessary to be classified according to the inflammatory state, those with high levels of baseline inflammation should receive combined treatment with anti-inflammatory agents like GZA.
Collapse
Affiliation(s)
- Zhi-Yong Cao
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China; Department of Psychiatry, The 904th Hospital of PLA, 55 North Heping Road, Changzhou, China
| | - Yun-Zi Liu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Jia-Mei Li
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yi-Ming Ruan
- Department of Health Statistics, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Wen-Jie Yan
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Shi-Yang Zhong
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Ting Zhang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Lin-Lin Liu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China; Department of Nursing, The 474th Hospital of PLA, 757 Beijing Road, Urumqi, China
| | - Ran Wu
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Bo Wang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Wei Wang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Xiao-Ying Bi
- Department of Neurology, Changhai Hospital, 168 Changhai Road, Shanghai, China
| | - Yun-Xia Wang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Wen-Jun Su
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China.
| | - Chun-Lei Jiang
- Department of Stress Medicine, Faculty of Psychology, Second Military Medical University, 800 Xiangyin Road, Shanghai, China.
| |
Collapse
|
10
|
Paudel YN, Angelopoulou E, Semple B, Piperi C, Othman I, Shaikh MF. Potential Neuroprotective Effect of the HMGB1 Inhibitor Glycyrrhizin in Neurological Disorders. ACS Chem Neurosci 2020; 11:485-500. [PMID: 31972087 DOI: 10.1021/acschemneuro.9b00640] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glycyrrhizin (glycyrrhizic acid), a bioactive triterpenoid saponin constituent of Glycyrrhiza glabra, is a traditional medicine possessing a plethora of pharmacological anti-inflammatory, antioxidant, antimicrobial, and antiaging properties. It is a known pharmacological inhibitor of high mobility group box 1 (HMGB1), a ubiquitous protein with proinflammatory cytokine-like activity. HMGB1 has been implicated in an array of inflammatory diseases when released extracellularly, mainly by activating intracellular signaling upon binding to the receptor for advanced glycation end products (RAGE) and toll-like receptor 4 (TLR4). HMGB1 neutralization strategies have demonstrated disease-modifying outcomes in several preclinical models of neurological disorders. Herein, we reveal the potential neuroprotective effects of glycyrrhizin against several neurological disorders. Emerging findings demonstrate the therapeutic potential of glycyrrhizin against several HMGB1-mediated pathological conditions including traumatic brain injury, neuroinflammation and associated conditions, epileptic seizures, Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Glycyrrhizin's effects in neurological disorders are mainly attributed to the attenuation of neuronal damage by inhibiting HMGB1 expression and translocation as well as by downregulating the expression of inflammatory cytokines. A large number of preclinical findings supports the notion that glycyrrhizin might be a promising therapeutic alternative to overcome the shortcomings of the mainstream therapeutic strategies against neurological disorders, mainly by halting disease progression. However, future research is warranted for a deeper exploration of the precise underlying molecular mechanism as well as for clinical translation.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Bridgette Semple
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Mohd Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| |
Collapse
|
11
|
Chen H, Guan B, Wang B, Pu H, Bai X, Chen X, Liu J, Li C, Qiu J, Yang D, Liu K, Wang Q, Qi S, Shen J. Glycyrrhizin Prevents Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke with Delayed Thrombolysis Through Targeting Peroxynitrite-Mediated HMGB1 Signaling. Transl Stroke Res 2019; 11:967-982. [PMID: 31872339 DOI: 10.1007/s12975-019-00772-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 11/17/2019] [Accepted: 12/09/2019] [Indexed: 01/22/2023]
Abstract
Peroxynitrite (ONOO-) and high mobility group box 1 protein (HMGB1) are important cytotoxic factors contributing to cerebral ischemia-reperfusion injury. However, the roles of ONOO- in mediating HMGB1 expression and its impacts on hemorrhagic transformation (HT) in ischemic brain injury with delayed t-PA treatment remain unclear. In the present study, we tested the hypothesis that ONOO- could directly mediate the activation and release of HMGB1 in ischemic brains with delayed t-PA treatment. With clinical studies, we found that plasma nitrotyrosine (NT, a surrogate marker of ONOO-) was positively correlated with HMGB1 level in acute ischemic stroke patients. Hemorrhagic transformation and t-PA-treated ischemic stroke patients had increased levels of nitrotyrosine and HMGB1 in plasma. In animal experiments, we found that FeTmPyP, a representative ONOO- decomposition catalyst (PDC), significantly reduced the expression of HMGB1 and its receptor TLR2, and inhibited MMP-9 activation, preserved collagen IV and tight junction claudin-5 in ischemic rat brains with delayed t-PA treatment. ONOO- donor SIN-1 directly induced expression of HMGB1 and its receptor TLR2 in naive rat brains in vivo and induced HMGB1 in brain microvascular endothelial b.End3 cells in vitro. Those results suggest that ONOO- could activate HMGB1/TLR2/MMP-9 signaling. We then addressed whether glycyrrhizin, a natural HMGB1 inhibitor, could inhibit ONOO- production and the antioxidant properties of glycyrrhizin contribute to the inhibition of HMGB1 and the neuroprotective effects on attenuating hemorrhagic transformation in ischemic stroke with delayed t-PA treatment. Glycyrrhizin treatment downregulated the expressions of NADPH oxidase p47 phox and p67 phox and iNOS, inhibited superoxide and ONOO- production, reduced the expression of HMGB1, TLR2, MMP-9, preserved type IV collagen and claudin-5 in ischemic brains. Furthermore, glycyrrhizin significantly decreased the mortality rate, attenuated hemorrhagic transformation, brain swelling, blood-brain barrier damage, neuronal apoptosis, and improved neurological outcomes in the ischemic stroke rat model with delayed t-PA treatment. In conclusion, peroxynitrite-mediated HMGB1/TLR2 signaling contributes to hemorrhagic transformation, and glycyrrhizin could be a potential adjuvant therapy to attenuate hemorrhagic transformation, possibly through inhibiting the ONOO-/HMGB1/TLR2 signaling cascades.
Collapse
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China.,Institute of Research and Innovation (HKU-SIRI), The University of Hong Kong-Shenzhen, Shenzhen, China
| | - Binghe Guan
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China
| | - Bin Wang
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Haiwei Pu
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Xiaoyu Bai
- Department of Chemistry, Morningside Laboratory for Chemical Biology, The University of Hong Kong, Hong Kong, SAR, China
| | - Xi Chen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China.,Department of Core Facility, The People's Hospital of Bao-an, Shenzhen, China
| | - Jihong Liu
- Department of Neurology, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Caiming Li
- Department of Neurology, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Jinhua Qiu
- Department of Neurology, Huizhou First Hospital, Huizhou, Guangdong, China
| | - Dan Yang
- Department of Chemistry, Morningside Laboratory for Chemical Biology, The University of Hong Kong, Hong Kong, SAR, China
| | - Kejian Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of New Mexico, Albuquerque, NM, 87131, USA
| | - Qi Wang
- Institution of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, SAR, China. .,Institute of Research and Innovation (HKU-SIRI), The University of Hong Kong-Shenzhen, Shenzhen, China. .,Institution of Clinical Pharmacology, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
12
|
Antifibrotic Effects of High-Mobility Group Box 1 Protein Inhibitor (Glycyrrhizin) on Keloid Fibroblasts and Keloid Spheroids through Reduction of Autophagy and Induction of Apoptosis. Int J Mol Sci 2019; 20:ijms20174134. [PMID: 31450620 PMCID: PMC6747471 DOI: 10.3390/ijms20174134] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/07/2023] Open
Abstract
Overabundance of extracellular matrix resulting from hyperproliferation of keloid fibroblasts (KFs) and dysregulation of apoptosis represents the main pathophysiology underlying keloids. High-mobility group box 1 (HMGB1) plays important roles in the regulation of cellular death. Suppression of HMGB1 inhibits autophagy while increasing apoptosis. Suppression of HMGB1 with glycyrrhizin has therapeutic benefits in fibrotic diseases. In this study, we explored the possible involvement of autophagy and HMGB1 as a cell death regulator in keloid pathogenesis. We have highlighted the potential utility of glycyrrhizin as an antifibrotic agent via regulation of the aberrant balance between autophagy and apoptosis in keloids. Higher HMGB1 expression and enhanced autophagy were observed in keloids. The proliferation of KFs was decreased following glycyrrhizin treatment. While apoptosis was enhanced in keloids after glycyrrhizin treatment, autophagy was significantly reduced. The expressions of ERK1/2, Akt, and NF-κB, were enhanced in HMGB1-teated fibroblasts, but decreased following glycyrrhizin treatment. The expression of extracellular matrix (ECM) components was reduced in glycyrrhizin-treated keloids. TGF-β, Smad2/3, ERK1/2, and HMGB1 were decreased in glycyrrhizin-treated keloids. Treatment with the autophagy inhibitor 3-MA resulted in a decrease of autophagy markers and collagen in the TGF-β-treated fibroblasts. The results indicated that autophagy plays an important role in the pathogenesis of keloids. Because glycyrrhizin appears to reduce ECM and downregulate autophagy in keloids, its potential use for treatment of keloids is indicated.
Collapse
|
13
|
Hisaoka-Nakashima K, Tomimura Y, Yoshii T, Ohata K, Takada N, Zhang FF, Nakamura Y, Liu K, Wake H, Nishibori M, Nakata Y, Morioka N. High-mobility group box 1-mediated microglial activation induces anxiodepressive-like behaviors in mice with neuropathic pain. Prog Neuropsychopharmacol Biol Psychiatry 2019; 92:347-362. [PMID: 30763674 DOI: 10.1016/j.pnpbp.2019.02.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/22/2019] [Accepted: 02/07/2019] [Indexed: 12/13/2022]
Abstract
Clinical evidence indicates that major depression is a common comorbidity of chronic pain, including neuropathic pain. However, the cellular basis for chronic pain-mediated major depression remains unclear. High-mobility group box 1 protein (HMGB1) has a key role in innate immune responses and appears to be have a role in mediating diverse disorders, including neuropathic pain and depression. The current study aimed to characterize neuropathic pain-induced changes in affect over time and to determine whether HMGB1 has a role in neuropathic pain-induced changes in affect. Neuropathic pain was induced by partial sciatic nerve ligation (PSNL) in mice. Anxiodepressive-like behaviors in mice were evaluated over 10 weeks, in the social interaction, forced swim, and novelty suppressed feeding tests. Mice developed anxiodepressive-like behavior 6 to 8 weeks after induction of neuropathy. Accompanying anxiodepressive-like behavior, increased HMGB1 protein and microglia activation were observed in frontal cortex at 8 weeks after PSNL. Intracerebroventricular administration of rHMGB1 in naïve mice induced anxiodepressive-like behavior and microglia activation. Blockage of HMGB1 in PSNL mice with glycyrrhizic acid (GZA) or anti-HMGB1 antibody reduced microglia activation and anxiodepressive-like behavior. These results indicate that PSNL-induced anxiodepressive-like behavior is likely mediated by HMGB1. Furthermore, the data indicate that inhibition of HMGB1-dependent microglia activation could be a strategy for the treatment of depression associated with neuropathic pain.
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
| | - Yoshiaki Tomimura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Toshiki Yoshii
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Kazuto Ohata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Naoki Takada
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Fang Fang Zhang
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan; Institute of Pharmacology, Tawishan Medical University, 619 Changcheng Road, Taian, Shandong 271016, China
| | - Yoki Nakamura
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Keyue Liu
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Hidenori Wake
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Masahiro Nishibori
- Department of Pharmacology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Shikata, Okayama, Japan
| | - Yoshihiro Nakata
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan
| | - Norimitsu Morioka
- Department of Pharmacology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Kasumi 1-2-3, Minami-ku, Hiroshima, Japan.
| |
Collapse
|
14
|
Chang HY, Chen SY, Wu CH, Lu CC, Yen GC. Glycyrrhizin Attenuates the Process of Epithelial-to-Mesenchymal Transition by Modulating HMGB1 Initiated Novel Signaling Pathway in Prostate Cancer Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3323-3332. [PMID: 30832473 DOI: 10.1021/acs.jafc.9b00251] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
High mobility group box 1 (HMGB1) is upregulated in nearly every tumor type. Importantly, clinical evidence also proposed that HMGB1 is particularly increased in metastatic prostate cancer patients. Besides, a growing number of studies highlighted that HMGB1 could be a successful therapeutic target for prostate cancer patients. Glycyrrhizin is a novel pharmacological inhibitor of HMGB1 that may repress prostate cancer metastasis. This research was aimed to investigate the effect of glycyrrhizin on inhibition of HMGB1-induced epithelial-to-mesenchymal transition (EMT), a key step of tumor metastasis, in prostate cancer cells. In this study, HMGB1 knock-downed DU145 prostate cancer cells were used. Silencing the HMGB1 gene expression triggered a change of cell morphology to a more epithelial-like shape, which was accompanied by a reduction of Cdc42/GSK-3β/Snail and induction of E-cadherin levels estimated by immunoblotting. Furthermore, HMGB1 facilitated cell migration and invasion via downstream signaling, whereas HMGB1 targeting by 10 mM ethyl pyruvate effectively inhibited EMT characteristics. Interestingly, cell migration capacity induced by HMGB1 in DU145 cells was abolished in a dose-dependent effect of 25-200 μM glycyrrhizin treatment. In conclusion, glycyrrhizin successfully inhibited HMGB1-induced EMT phenomenon, which suggested that glycyrrhizin may serves as a therapeutic agent for metastatic prostate cancer.
Collapse
Affiliation(s)
| | | | - Chi-Hao Wu
- Department of Human Development and Family Studies , National Taiwan Normal University , 162, Section 1, Heping East Road , Taipei City 106 , Taiwan
| | - Chi-Cheng Lu
- Department of Sport Performance , National Taiwan University of Sport , 16, Sec. 1, Shuang-Shih Road , Taichung City 40404 , Taiwan
| | | |
Collapse
|
15
|
Paudel YN, Semple BD, Jones NC, Othman I, Shaikh MF. High mobility group box 1 (HMGB1) as a novel frontier in epileptogenesis: from pathogenesis to therapeutic approaches. J Neurochem 2019; 151:542-557. [DOI: 10.1111/jnc.14663] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/02/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory Jeffrey Cheah School of Medicine and Health Sciences Monash University Malaysia Bandar Sunway Selangor Malaysia
| | - Bridgette D. Semple
- Department of Neuroscience Central Clinical School Monash University The Alfred Hospital Melbourne Australia
- Department of Medicine (Royal Melbourne Hospital) The University of Melbourne Royal Parade Parkville Victoria Australia
| | - Nigel C. Jones
- Department of Neuroscience Central Clinical School Monash University The Alfred Hospital Melbourne Australia
- Department of Medicine (Royal Melbourne Hospital) The University of Melbourne Royal Parade Parkville Victoria Australia
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory Jeffrey Cheah School of Medicine and Health Sciences Monash University Malaysia Bandar Sunway Selangor Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory Jeffrey Cheah School of Medicine and Health Sciences Monash University Malaysia Bandar Sunway Selangor Malaysia
| |
Collapse
|
16
|
Wang Y, Zhang Y, Peng G, Han X. Glycyrrhizin ameliorates atopic dermatitis-like symptoms through inhibition of HMGB1. Int Immunopharmacol 2018; 60:9-17. [PMID: 29702284 DOI: 10.1016/j.intimp.2018.04.029] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/13/2018] [Accepted: 04/13/2018] [Indexed: 12/29/2022]
Abstract
Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disease prevalent worldwide. This study investigated the effects of glycyrrhizin, an extract of licorice root, on the well-established model of 2,4-dinitrochlorobenzene-induced AD-like symptoms in mice. The severity of dermatitis, histopathological changes, serum IgE levels, changes in expression of high-mobility group box 1 (HMGB1), the receptor for advanced glycation end products (RAGE), nuclear factor (NF)-κB and inflammatory cytokines were evaluated. Treatment with glycyrrhizin inhibited the HMGB1 signaling cascade and ameliorated the symptoms of AD. Furthermore, in an in vitro study, the expression of RAGE was detected in a mouse mast cell line, P815 cells, and rmHMGB1 was found to be a potent inducer of mast cell activation by increasing Ca2+ influx, upregulating the CD117 and activating NF-κB signaling; these effects were also inhibited by glycyrrhizin. These findings implicate HMGB1 in the pathogenesis of AD and suggest that GL could be an effective therapeutic approach for cutaneous inflammation.
Collapse
Affiliation(s)
- Ying Wang
- Department of Dermatology, Shengjing Hospital of China Medical University, Heping District, Shenyang, Liaoning 110004, China
| | - Yue Zhang
- Department of Dermatology, Shengjing Hospital of China Medical University, Heping District, Shenyang, Liaoning 110004, China
| | - Ge Peng
- Department of Dermatology, Shengjing Hospital of China Medical University, Heping District, Shenyang, Liaoning 110004, China
| | - Xiuping Han
- Department of Dermatology, Shengjing Hospital of China Medical University, Heping District, Shenyang, Liaoning 110004, China.
| |
Collapse
|
17
|
Wang B, Lian YJ, Dong X, Peng W, Liu LL, Su WJ, Gong H, Zhang T, Jiang CL, Li JS, Wang YX. Glycyrrhizic acid ameliorates the kynurenine pathway in association with its antidepressant effect. Behav Brain Res 2018; 353:250-257. [PMID: 29366745 DOI: 10.1016/j.bbr.2018.01.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 12/20/2022]
Abstract
Our previous study implied the role of central high mobility group box 1 (HMGB1) in lipopolysaccharide (LPS)-induced depressive-like behaviors that could partially abrogate by glycyrrhizic acid (GZA). Here, we considered the potential mechanism underlying GZA ameliorating chronic stress-induced depression both in vivo and in vitro. Depression model was established with the 4-week chronic unpredictable mild stress (CUMS) mice. Sucrose preference test, tail suspension test and open field test were performed to reflect depressive-like behaviors. Enzyme activity of indoleamine-2,3-dioxygenase (IDO) was recorded with the ratio of kynurenine (KYN) / tryptophan (Trp). Transcription of gene was evaluated by RT-PCR. Along with depressive-like behaviors, IDO, the rate-limiting enzyme of the kynurenine pathway (KP), was upregulated at the level of mRNA expression, and enzyme activity was also elevated in stressed hippocampi and LPS/HMGB1-treated hippocampus slices. Treatment of mice with GZA, the inhibitor of HMGB1, prevented the activated enzymes in KP and the development of depressive-like behaviors. These experiments demonstrate that GZA may restrain HMGB1 thus improving chronic stress-induced depressive behavior through regulating KP.
Collapse
Affiliation(s)
- Bo Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Yong-Jie Lian
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Xin Dong
- School of Pharmacy, Second Military Medical University, Shanghai 200433, China
| | - Wei Peng
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Psychiatry, The 92nd Hospital of PLA, Nanping 353000, China
| | - Lin-Lin Liu
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China; Department of Nursing, The 474th Hospital of PLA, Urumqi 830012, China
| | - Wen-Jun Su
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Hong Gong
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Ting Zhang
- Department of Navy Medicine, Second Military Medical University, Shanghai 200433, China
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China
| | - Jia-Si Li
- Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Yun-Xia Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
18
|
Rosenberg JH, Rai V, Dilisio MF, Agrawal DK. Damage-associated molecular patterns in the pathogenesis of osteoarthritis: potentially novel therapeutic targets. Mol Cell Biochem 2017; 434:171-179. [PMID: 28474284 DOI: 10.1007/s11010-017-3047-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
Abstract
Osteoarthritis (OA) is a chronic disease that degrades the joints and is often associated with increasing age and obesity. The two most common sites of OA in adults are the knee and hip joints. Increased mechanical stress on the joint from obesity can cause the articular cartilage to degrade and release damage-associated molecular patterns (DAMPs). These DAMPs are involved in various molecular pathways that interact with nuclear factor-kappa B and result in the transcription of inflammatory cytokines and activation of matrix metalloproteinases that progressively destroy cartilage. This review focuses on the interactions and contribution to the pathogenesis and progression of OA through the DAMPs: high-mobility group box 1 (HMGB-1), the receptor for advanced glycation end-products (RAGE), the alarmin proteins S100A8 and S100A9, and heparan sulfate. HMGB-1 is released from damaged or necrotic cells and interacts with toll-like receptors (TLRs) and RAGE to induce inflammatory signals, as well as behave as an inflammatory cytokine to activate innate immune cells. RAGE interacts with HMGB-1, advanced glycation end-products, and innate immune cells to increase local inflammation. The alarmin proteins are released following cell damage and interact through TLRs to increase local inflammation and cartilage degradation. Heparan sulfate has been shown to facilitate the binding of HMGB-1 to RAGE and could play a role in the progression of OA. Targeting these DAMPs may be the potential therapeutic strategies for the treatment of OA.
Collapse
Affiliation(s)
- John H Rosenberg
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA
| | - Matthew F Dilisio
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.,Department of Orthopedic Surgery, Creighton University School of Medicine, Omaha, NE, 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, CRISS II Room 510, 2500 California Plaza, Omaha, NE, 68178, USA.
| |
Collapse
|
19
|
Glycyrrhizin ameliorates experimental colitis through attenuating interleukin-17-producing T cell responses via regulating antigen-presenting cells. Immunol Res 2017; 65:666-680. [DOI: 10.1007/s12026-017-8894-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
20
|
Venereau E, De Leo F, Mezzapelle R, Careccia G, Musco G, Bianchi ME. HMGB1 as biomarker and drug target. Pharmacol Res 2016; 111:534-544. [PMID: 27378565 DOI: 10.1016/j.phrs.2016.06.031] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 06/29/2016] [Accepted: 06/30/2016] [Indexed: 12/27/2022]
Abstract
High Mobility Group Box 1 protein was discovered as a nuclear protein, but it has a "second life" outside the cell where it acts as a damage-associated molecular pattern. HMGB1 is passively released or actively secreted in a number of diseases, including trauma, chronic inflammatory disorders, autoimmune diseases and cancer. Extracellular HMGB1 triggers and sustains the inflammatory response by inducing cytokine release and by recruiting leucocytes. These characteristics make extracellular HMGB1 a key molecular target in multiple diseases. A number of strategies have been used to prevent HMGB1 release or to inhibit its activities. Current pharmacological strategies include antibodies, peptides, decoy receptors and small molecules. Noteworthy, salicylic acid, a metabolite of aspirin, has been recently found to inhibit HMGB1. HMGB1 undergoes extensive post-translational modifications, in particular acetylation and oxidation, which modulate its functions. Notably, high levels of serum HMGB1, in particular of the hyper-acetylated and disulfide isoforms, are sensitive disease biomarkers and are associated with different disease stages. In the future, the development of isoform-specific HMGB1 inhibitors may potentiate and fine-tune the pharmacological control of inflammation. We review here the current therapeutic strategies targeting HMGB1, in particular the emerging and relatively unexplored small molecules-based approach.
Collapse
Affiliation(s)
- Emilie Venereau
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; HMGBiotech s.r.l., Milan, Italy
| | - Federica De Leo
- Biomolecular NMR Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosanna Mezzapelle
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Careccia
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; University of Milano Bicocca, Milan, Italy
| | - Giovanna Musco
- Biomolecular NMR Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco E Bianchi
- Chromatin Dynamics Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; San Raffaele University, Milan, Italy.
| |
Collapse
|
21
|
Liu J, Zhang BL, Sun CL, Wang J, Li S, Wang JF. High mobility group box1 protein is involved in acute inflammation induced by Clostridium difficile toxin A. Acta Biochim Biophys Sin (Shanghai) 2016; 48:554-62. [PMID: 27151296 DOI: 10.1093/abbs/gmw038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/31/2016] [Indexed: 12/31/2022] Open
Abstract
High mobility group box1 (HMGB1), as a damage-associated inflammatory factor, contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases. In this study, we explored the role of HMGB1 in CDI (Clostridium difficile infection) by in vivo and in vitro experiments. Our results showed that HMGB1 might play an important role in the acute inflammatory responses to C. difficile toxin A (TcdA), affect early inflammatory factors, and induce inflammation via the HMGB1-TLR4 pathway. Our study provides the essential information for better understanding the molecular mechanisms of CDI and the potential new therapeutic strategies for the treatment of this infection.
Collapse
Affiliation(s)
- Ji Liu
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Bei-Lei Zhang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Chun-Li Sun
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- Shenzhen Huada Gene Research Institute, Shenzhen 518083, China
| | - Shan Li
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China Guangdong Province Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ju-Fang Wang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
22
|
Wu TY, Liu L, Zhang W, Zhang Y, Liu YZ, Shen XL, Gong H, Yang YY, Bi XY, Jiang CL, Wang YX. High-mobility group box-1 was released actively and involved in LPS induced depressive-like behavior. J Psychiatr Res 2015; 64:99-106. [PMID: 25795092 DOI: 10.1016/j.jpsychires.2015.02.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 02/20/2015] [Accepted: 02/20/2015] [Indexed: 01/01/2023]
Abstract
Depression disorder is a common mental illness, of which the pathogenesis is not well understood. Studies suggest that immunity imbalance and up-regulation of pro-inflammatory cytokines may be associated with the pathogenesis of depression. High-mobility group box 1 protein (HMGB1) has gained much attention as an important player in innate immune responses and an modulating factor in several inflammatory diseases. Here we sought to explore the role of HMGB1 in the development of depression. Depression model was established with low dose of lipopolysaccharide (LPS) administration. Depressive behavior was reflected with increased immobility time in tail suspension test. Accompanying with depressive-like behavior, translocation of HMGB1 from nuclei to cytoplasm was observed by immunofluorescence assays. Meanwhile, no significant necrosis was observed evaluated by hematoxylin-eosin staining. These data indicated that HMGB1 was released actively in the central nervous system. In addition, treating the mice with human recombinant HMGB1 (rHMGB1) could induce the development of depressive-like behavior. Blockage of HMGB1 with GZA abrogated the depressive-like behavior induced by LPS or rHMGB1. These results implicated that HMGB1 was involved in LPS-induced depressive-like behavior.
Collapse
Affiliation(s)
- Teng-Yun Wu
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Lei Liu
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Wei Zhang
- Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | - Yi Zhang
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Yun-Zi Liu
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Xiao-Liang Shen
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Hong Gong
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Yuan-Yuan Yang
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Xiao-Ying Bi
- Department of Neurology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, PR China.
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| | - Yun-Xia Wang
- Lab of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, Shanghai, PR China.
| |
Collapse
|
23
|
Kim YM, Kim HJ, Chang KC. Glycyrrhizin reduces HMGB1 secretion in lipopolysaccharide-activated RAW 264.7 cells and endotoxemic mice by p38/Nrf2-dependent induction of HO-1. Int Immunopharmacol 2015; 26:112-8. [PMID: 25812767 DOI: 10.1016/j.intimp.2015.03.014] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/04/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
High mobility group box 1 (HMGB1) is now recognized as a late mediator of sepsis. Although glycyrrhizin was known as inhibitor of HMGB1, it is not yet clear underlying mechanism(s). We found that glycyrrhizin activates translocation of Nrf2 from cytosol to nucleus and induces heme oxygenase (HO)-1 expression in RAW 264.7 cells. In addition, deletion of Nrf2 by siRNA significantly reduced mRNA expression of NQO1 and HO-1 suggesting that glycyrrhizin targets Nrf2 gene. The expression of iNOS protein and release of HMGB1 in LPS activated cells were significantly reduced by glycyrrhizin and cells transfected with mouse HO-1 expression vector. The p38MAPK inhibitor (SB203580) but not JNK inhibitor (SP600125) or ERK inhibitor (PD98059) significantly inhibited HO-1 induction by glycyrrhizin, which was confirmed by showing that siP38 transfected cells significantly reduced HO-1 induction. Pretreatment with SB203580 significantly reversed the expression of iNOS and release of NO and HMGB1 in LPS-activated cells. Most importantly, administration of glycyrrhizin (200mg/kg, i.p) significantly reduced hepatic injury and serum HMGB1 in a ZnPPIX-sensitive manner. Thus, it is concluded that glycyrrhizin reduces HMGB1 secretion in lipopolysaccharide-activated RAW 264.7 cells and endotoxemic mice by p38/Nrf2-dependent induction of HO-1.
Collapse
Affiliation(s)
- Young Min Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea
| | - Ki Churl Chang
- Department of Pharmacology, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-751, Republic of Korea.
| |
Collapse
|
24
|
PGE2 receptor agonist misoprostol protects brain against intracerebral hemorrhage in mice. Neurobiol Aging 2015; 36:1439-50. [PMID: 25623334 DOI: 10.1016/j.neurobiolaging.2014.12.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 12/16/2014] [Accepted: 12/26/2014] [Indexed: 11/23/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke. Misoprostol, a synthetic prostaglandin E1 (PGE1) analog and PGE2 receptor agonist, has shown protection against cerebral ischemia. In this study, we tested the efficacy of misoprostol in the 12-month-old mice subjected to 1 of 2 complementary ICH models, the collagenase model (primary study) and blood model (secondary study, performed in an independent laboratory). We also investigated its potential mechanism of action. Misoprostol posttreatment decreased brain lesion volume, edema, and brain atrophy and improved long-term functional outcomes. In the collagenase-induced ICH model, misoprostol decreased cellular inflammatory response; attenuated oxidative brain damage and gelatinolytic activity; and decreased high-mobility group box 1 (HMGB1) expression, Src kinase activity, and interleukin-1β expression without affecting cyclooxygenase-2 expression. Furthermore, HMGB1 inhibition with glycyrrhizin decreased Src kinase activity, gelatinolytic activity, neuronal death, and brain lesion volume. Src kinase inhibition with 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d]pyrimidine (PP2) decreased gelatinolytic activity and brain edema and improved neurologic function but did not decrease HMGB1 protein level. These results indicate that misoprostol protects brain against ICH injury through mechanisms that may involve the HMGB1, Src kinase, and matrix metalloproteinase-2/9 pathways.
Collapse
|
25
|
Zhang CC, Gdynia G, Ehemann V, Roth W. The HMGB1 protein sensitizes colon carcinoma cells to cell death triggered by pro-apoptotic agents. Int J Oncol 2014; 46:667-76. [PMID: 25434832 DOI: 10.3892/ijo.2014.2785] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 11/06/2014] [Indexed: 11/06/2022] Open
Abstract
The HMGB1 protein has multiple functions in tumor biology and can act both as a transcription factor and as a cytokine. HMGB1 is released during cell death, and in our previous studies we demonstrated that HMGB1 induces a distinct, necrosis-like cell death in glioblastoma. In epithelial malignant tumors such as colorectal cancer (CRC), the HMGB1-dependent effects show cross-talk with apoptotic signal transduction. Treatment of CRC cells with low concentrations of recombinant HMGB1 results in dose-dependent cytotoxicity which is morphologically characterized by the formation of giant mitochondria and does not share features of apoptosis. HMGB1-triggered cell death is associated with intracellular ROS release, and overexpression of Bcl-2 blocks both the increase of ROS as well as HMGB1-dependent cell death. Importantly, treatment with recombinant HMGB1 or overexpression of endogenous HMGB1 strongly sensitizes CRC cells to the cytotoxic activity of the pro-apoptotic death ligand TRAIL as well as the small molecule Bcl-2 family inhibitor ABT‑737. Moreover, treatment of CRC cells with TRAIL or ABT‑737 induces a release of endogenous HMGB1 into the extracellular space, and preincubation with glycyrrhizin, an HMGB1 inhibitor, significantly inhibits induction of cell death by TRAIL and ABT‑737, suggesting that HMGB1 functionally contributes to the execution of cell death triggered by pro-apoptotic agents. Finally, we investigated the expression of HMGB1 in human CRC tumor samples and found that loss of HMGB1 expression is associated with a more aggressive phenotype and a more advanced stage of disease in patients with CRC. Altogether, our findings demonstrate a functional link between cytotoxic signaling cascades triggered by HMGB1 and pro-apoptotic agents leading to an HMGB1-dependent sensitization to CRC cell death. Thus, a further evaluation of recombinant HMGB1 as part of an experimental combination treatment of CRC seems warranted.
Collapse
Affiliation(s)
- Chengcheng Christine Zhang
- Molecular Tumor Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Georg Gdynia
- Molecular Tumor Pathology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | - Volker Ehemann
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| | - Wilfried Roth
- Institute of Pathology, University of Heidelberg, Im Neuenheimer Feld 224, D-69120 Heidelberg, Germany
| |
Collapse
|
26
|
Galbiati V, Papale A, Galli CL, Marinovich M, Corsini E. Role of ROS and HMGB1 in Contact Allergen–Induced IL-18 Production in Human Keratinocytes. J Invest Dermatol 2014; 134:2719-2727. [DOI: 10.1038/jid.2014.203] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 03/18/2014] [Accepted: 04/07/2014] [Indexed: 12/25/2022]
|
27
|
Sun Q, Wu W, Hu YC, Li H, Zhang D, Li S, Li W, Li WD, Ma B, Zhu JH, Zhou ML, Hang CH. Early release of high-mobility group box 1 (HMGB1) from neurons in experimental subarachnoid hemorrhage in vivo and in vitro. J Neuroinflammation 2014; 11:106. [PMID: 24924349 PMCID: PMC4107626 DOI: 10.1186/1742-2094-11-106] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 05/29/2014] [Indexed: 12/25/2022] Open
Abstract
Background Translocation of high-mobility group box 1 (HMGB1) from nucleus could trigger inflammation. Extracellular HMGB1 up-regulates inflammatory response in sepsis as a late mediator. However, little was known about its role in subarachnoid hemorrhage-inducible inflammation, especially in the early stage. This study aims to identify whether HMGB1 translocation occurred early after SAH and also to clarify the potential role of HMGB1 in brain injury following SAH. Methods Sprague-Dawley (SD) rats were randomly divided into sham group and SAH groups at 2 h, 12 h and on day 1, day 2. SAH groups suffered experimental subarachnoid hemorrhage by injection of 0.3 ml autoblood into the pre-chiasmatic cistern. Rats injected by recombinant HMGB1(rHMGB1) solution were divided into four groups according to different time points. Cultured neurons were assigned into control group and four hemoglobin (Hb) incubated groups. Mixed glial cells were cultured and stimulated in medium from neurons incubated by Hb. HMGB1 expression is measured by western blot analysis, real-time polymerase chain reaction (PCR), immunohistochemistry and immunofluorescence. Downstream nuclear factor kappa B (NF-κB) subunit P65 and inflammatory factor Interleukin 1β (IL-1β) were measured by western blot and real-time PCR, respectively. Brain injury was evaluated by cleaved caspase-3 staining. Results Our results demonstrated HMGB1 translocation occurred as early as 2 h after experimental SAH with mRNA and protein level increased. Immunohistochemistry and immunofluorescence results indicated cytosolic HMGB1 was mainly located in neurons while translocated HMGB1 could also be found in some microglia. After subarachnoid injection of rHMGB1, NF-κB, downstream inflammatory response and cleaved caspase-3 were up-regulated in the cortex compared to the saline control group. In-vitro, after Hb incubation, HMGB1 was also rapidly released from neurons to medium. Incubation with medium from neurons up-regulated IL-1β in mixed glial cells. This effect could be inhibited by HMGB1 specific inhibitor glycyrrhizic acid (GA) treatment. Conclusion HMGB1 was released from neurons early after SAH onset and might trigger inflammation as an upstream inflammatory mediator. Extracellular HMGB1 contributed to the brain injury after SAH. These results might have important implications during the administration of specific HMGB1 antagonists early in order to prevent or reduce inflammatory response following SAH.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Chun-Hua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
28
|
Sattler S, Ling GS, Xu D, Hussaarts L, Romaine A, Zhao H, Fossati-Jimack L, Malik T, Cook HT, Botto M, Lau YL, Smits HH, Liew FY, Huang FP. IL-10-producing regulatory B cells induced by IL-33 (Breg(IL-33)) effectively attenuate mucosal inflammatory responses in the gut. J Autoimmun 2014; 50:107-22. [PMID: 24491821 PMCID: PMC4012142 DOI: 10.1016/j.jaut.2014.01.032] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 12/13/2013] [Accepted: 01/13/2014] [Indexed: 12/02/2022]
Abstract
Regulatory B cells (Breg) have attracted increasing attention for their roles in maintaining peripheral tolerance. Interleukin 33 (IL-33) is a recently identified IL-1 family member, which leads a double-life with both pro- and anti-inflammatory properties. We report here that peritoneal injection of IL-33 exacerbated inflammatory bowel disease in IL-10-deficient (IL-10(-/-)) mice, whereas IL-33-treated IL-10-sufficient (wild type) mice were protected from the disease induction. A phenotypically unconventional subset(s) (CD19(+)CD25(+)CD1d(hi)IgM(hi)CD5(-)CD23(-)Tim-1(-)) of IL-10 producing Breg-like cells (Breg(IL-33)) was identified responsible for the protection. We demonstrated further that Breg(IL-33) isolated from these mice could suppress immune effector cell expansion and functions and, upon adoptive transfer, effectively blocked the development of spontaneous colitis in IL-10(-/-) mice. Our findings indicate an essential protective role, hence therapeutic potential, of Breg(IL-33) against mucosal inflammatory disorders in the gut.
Collapse
Affiliation(s)
- Susanne Sattler
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - Guang-Sheng Ling
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK; Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China; Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Damo Xu
- Division of Immunology, Infection and Inflammation, University of Glasgow, Glasgow, UK
| | - Leonie Hussaarts
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Romaine
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - Hongzhi Zhao
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK; Department of Hepatobiliary Surgery, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Liliane Fossati-Jimack
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - Talat Malik
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - H Terence Cook
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - Marina Botto
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK
| | - Yu-Lung Lau
- Department of Paediatrics & Adolescent Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Foo Y Liew
- Division of Immunology, Infection and Inflammation, University of Glasgow, Glasgow, UK
| | - Fang-Ping Huang
- Department of Medicine, Division of Immunology & Inflammation, Imperial College, London, UK; Department of Pathology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China.
| |
Collapse
|
29
|
Srinivasan M, Banerjee S, Palmer A, Zheng G, Chen A, Bosland MC, Kajdacsy-Balla A, Kalyanasundaram R, Munirathinam G. HMGB1 in hormone-related cancer: a potential therapeutic target. Discov Oncol 2014; 5:127-39. [PMID: 24718937 DOI: 10.1007/s12672-014-0175-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Accepted: 03/20/2014] [Indexed: 02/06/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a dynamic nuclear protein participating in transcription, chromatin remodelling, and DNA recombination and repair processes. Accumulating evidence indicates that its function now extends beyond the nucleus, notably its extracellular role in inflammation. HMGB1 is implicated as a late mediator of sepsis and is also believed to promote atherosclerosis and other inflammatory diseases such as rheumatoid arthritis and systemic lupus erythematosus. Interestingly, deregulation of HMGB1 is shown to be associated with the hallmarks of cancer development. Moreover, several clinical studies have shown that HMGB1 is a promising biomarker for a variety of cancer types. In this review, we provide novel insights into the role and mechanisms of HMGB1, in particular, to hormone-related cancers and its potential to serve as a therapeutic target.
Collapse
Affiliation(s)
- Madhuwanti Srinivasan
- Department of Biomedical Sciences, University of Illinois, College of Medicine, 1601 Parkview Ave, Rockford, IL, 61107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Whitman BA, Knapp DJ, Werner DF, Crews FT, Breese GR. The cytokine mRNA increase induced by withdrawal from chronic ethanol in the sterile environment of brain is mediated by CRF and HMGB1 release. Alcohol Clin Exp Res 2013; 37:2086-97. [PMID: 23895427 PMCID: PMC3815509 DOI: 10.1111/acer.12189] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 05/02/2013] [Indexed: 01/03/2023]
Abstract
BACKGROUND Many neurobiological factors may initiate and sustain alcoholism. Recently, dysregulation of the neuroimmune system by chronic ethanol (CE) has implicated Toll-like receptor 4 (TLR4) activation. Even though TLR4s are linked to CE initiation of brain cytokine mRNAs, the means by which CE influences neuroimmune signaling in brain in the absence of infection remains uncertain. Therefore, the hypothesis is tested that release of an endogenous TLR4 agonist, high-mobility group box 1 (HMGB1) and/or corticotropin-releasing factor (CRF) during CE withdrawal are responsible for CE protocols increasing cytokine mRNAs. METHODS Acute ethanol (EtOH; 2.75 g/kg) and acute lipopolysaccharide (LPS; 250 μg/kg) dosing on cytokine mRNAs are first compared. Then, the effects of chronic LPS exposure (250 μg/kg for 10 days) on cytokine mRNAs are compared with changes induced by CE protocols (15 days of continuous 7% EtOH diet [CE protocol] or 3 intermittent 5-day cycles of 7% EtOH diet [CIE protocol]). Additionally, TLR4, HMGB1, and downstream effector mRNAs are assessed after CE, CIE, and chronic LPS. To test whether HMGB1 and/or CRF support the CE withdrawal increase in cytokine mRNAs, the HMGB1 antagonists, glycyrrhizin and ethyl pyruvate, and a CRF1 receptor antagonist (CRF1RA) are administered during 24 hours of CE withdrawal. RESULTS While cytokine mRNAs were not increased following acute EtOH, acute LPS increased all cytokine mRNAs 4 hours after injection. CE produced no change in cytokine mRNAs prior to CE removal; however, the CE and CIE protocols increased cytokine mRNAs by 24 hours after withdrawal. In contrast, chronic LPS produced no cytokine mRNA changes 24 hours after LPS dosing. TLR4 mRNA was elevated 24 hours following both CE protocols and chronic LPS exposure. While chronic LPS had no effect on HMGB1 mRNA, withdrawal from CE protocols significantly elevated HMGB1 mRNA. Systemic administration of HMGB1 antagonists or a CRF1RA significantly reduced the cytokine mRNA increase following CE withdrawal. The CRF1RA and the HMGB1 antagonist, ethyl pyruvate, also reduced the HMGB1 mRNA increase that followed CE withdrawal. CONCLUSIONS By blocking HMGB1 or CRF action during CE withdrawal, evidence is provided that HMGB1 and CRF release are critical for the CE withdrawal induction of selected brain cytokine mRNAs. Consequently, these results clarify a means by which withdrawal from CE exposure activates neuroimmune function in the sterile milieu of brain.
Collapse
Affiliation(s)
- Buddy A Whitman
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | |
Collapse
|
31
|
Sun Q, Wang F, Li W, Li W, Hu YC, Li S, Zhu JH, Zhou M, Hang CH. Glycyrrhizic acid confers neuroprotection after subarachnoid hemorrhage via inhibition of high mobility group box-1 protein: A hypothesis for novel therapy of subarachnoid hemorrhage. Med Hypotheses 2013; 81:681-5. [DOI: 10.1016/j.mehy.2013.07.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/01/2013] [Accepted: 07/13/2013] [Indexed: 12/01/2022]
|
32
|
Mencucci R, Favuzza E, Menchini U. Assessment of the tolerability profile of an ophthalmic solution of 5% glycyrrhizin and copolymer PEG/PPG on healthy volunteers and evaluation of its efficacy in the treatment of moderate to severe blepharitis. Clin Ophthalmol 2013; 7:1403-10. [PMID: 23874081 PMCID: PMC3712082 DOI: 10.2147/opth.s47657] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose To evaluate the tolerability on healthy volunteers and the efficacy on subjects affected by chronic moderate/severe blepharitis of a 5% glycyrrhizin and copolymer poly(ethylene glycol)/poly(propylene glycol)(PEG/PPG) ophthalmic solution. Methods The study was a randomized, controlled, open label, intra-patient monocentric study. It consisted of two different phases, the assessment of tolerability phase on 20 healthy volunteers, and the evaluation of the efficacy on 21 subjects affected by chronic moderate/severe blepharitis; the treatment period was 2 weeks, followed by 1-week of follow-up. In the efficacy phase, in both eyes, eyelid hygiene was also performed. At day 0, 3, 7, 14, and 21 a complete ophthalmological examination was performed. In the tolerability phase, signs of clinical toxicity were recorded and subject-reported symptoms were collected using a questionnaire. In the efficacy phase, global signs and symptoms of blepharitis scores were collected using standardized photographic scales and questionnaire. The statistical analysis was performed using the Wilcoxon signed-rank test. Results No ocular signs of drug toxicity were reported. During the treatment period for tolerability phase, there were statistically significant higher scores of tearing and ocular discomfort in the tolerability study group versus the tolerability control group. In the efficacy phase, differences between global scores of the two groups were statistically significant at day 0 (score of the efficacy study group was higher than the efficacy control group; P = 0.005) and at day 21 (score of the efficacy study group was lower than the efficacy control group (P ≤ 0.001).The difference of global scores at day 3, 7, 14, and 21 versus day 0 was statistically significant in both groups. No serious adverse events occurred. Conclusion The 5% glycyrrhizin ophthalmic solution was well tolerated in healthy volunteers and in patients with chronic moderate/severe blepharitis, and in association with eyelid hygiene showed good clinical anti-inflammatory activity that lasted after instillation suspension.
Collapse
Affiliation(s)
- Rita Mencucci
- Department of Surgery and Translational Medicine - Eye Clinic, University of Florence, Florence, Italy
| | | | | |
Collapse
|
33
|
Seol D, McCabe DJ, Choe H, Zheng H, Yu Y, Jang K, Walter MW, Lehman AD, Ding L, Buckwalter JA, Martin JA. Chondrogenic progenitor cells respond to cartilage injury. ACTA ACUST UNITED AC 2013; 64:3626-3637. [PMID: 22777600 DOI: 10.1002/art.34613] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Hypocellularity resulting from chondrocyte death in the aftermath of mechanical injury is thought to contribute to posttraumatic osteoarthritis. However, we observed that nonviable areas in cartilage injured by blunt impact were repopulated within 7-14 days by cells that appeared to migrate from the surrounding matrix. The aim of this study was to assess our hypothesis that the migrating cell population included chondrogenic progenitor cells that were drawn to injured cartilage by alarmins. METHODS Osteochondral explants obtained from mature cattle were injured by blunt impact or scratching, resulting in localized chondrocyte death. Injured sites were serially imaged by confocal microscopy, and migrating cells were evaluated for chondrogenic progenitor characteristics. Chemotaxis assays were used to measure the responses to chemokines, injury-conditioned medium, dead cell debris, and high mobility group box chromosomal protein 1 (HMGB-1). RESULTS Migrating cells were highly clonogenic and multipotent and expressed markers associated with chondrogenic progenitor cells. Compared with chondrocytes, these cells overexpressed genes involved in proliferation and migration and underexpressed cartilage matrix genes. They were more active than chondrocytes in chemotaxis assays and responded to cell lysates, conditioned medium, and HMGB-1. Glycyrrhizin, a chelator of HMGB-1 and a blocking antibody to receptor for advanced glycation end products (RAGE), inhibited responses to cell debris and conditioned medium and reduced the numbers of migrating cells on injured explants. CONCLUSION Injuries that caused chondrocyte death stimulated the emergence and homing of chondrogenic progenitor cells, in part via HMGB-1 release and RAGE-mediated chemotaxis. Their repopulation of the matrix could promote the repair of chondral damage that might otherwise contribute to progressive cartilage loss.
Collapse
Affiliation(s)
- Dongrim Seol
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Daniel J McCabe
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Hyeonghun Choe
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Hongjun Zheng
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Yin Yu
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Keewoong Jang
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Morgan W Walter
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Abigail D Lehman
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Lei Ding
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| | - Joseph A Buckwalter
- Joseph A. Buckwalter, MD: University of Iowa and VA Medical Center, Iowa City, Iowa
| | - James A Martin
- Dongrim Seol, PhD, Daniel J. McCabe, BS, Hyeonghun Choe, ME, Hongjun Zheng, PhD, Yin Yu, BM, Keewoong Jang, MS, Morgan W. Walter, BS, Abigail D. Lehman, BS, Lei Ding, PhD, James A. Martin, PhD: University of Iowa, Iowa City
| |
Collapse
|
34
|
Lee YM, Kim J, Jo K, Shin SD, Kim CS, Sohn EJ, Kim SG, Kim JS. Ethyl pyruvate inhibits retinal pathogenic neovascularization by downregulating HMGB1 expression. J Diabetes Res 2013; 2013:245271. [PMID: 24371837 PMCID: PMC3858882 DOI: 10.1155/2013/245271] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 10/18/2013] [Accepted: 11/08/2013] [Indexed: 02/07/2023] Open
Abstract
Retinal pathogenic angiogenesis in the eyes is a causative factor in retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration. This study was designed to examine the pathogenic role of the high-mobility group box-1 (HMGB1) protein and the inhibitory effect of ethyl pyruvate (EP), a well-known antioxidant substance, in retinal pathogenic angiogenesis in mice with oxygen-induced retinopathy (OIR), one of the animal models of proliferative ischemic retinopathy. The OIR mouse model was used for our in vivo studies. The mice were exposed to 75% oxygen from postnatal day 7 (P7) to P11, after which the mice were brought to room air and intraperitoneally injected with EP (50 mg/kg, or 100 mg/kg) for five days. At P17, the mice were perfused with fluorescein isothiocyanate-dextran, and flat-mounted retinas were used to measure nonperfused and neovascular tufts. In OIR mice, an intraperitoneal injection of EP reduced the nonperfused retinal area in the treatment group and significantly reduced the retinal neovascular tufts. In addition, EP inhibited the overexpression of HMGB1 in the retinas of OIR mice. These data suggest that EP could serve as an innovative pharmaceutical agent to prevent retinal neovascularization through inhibiting HMGB1 expression.
Collapse
Affiliation(s)
- Yun Mi Lee
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Junghyun Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Kyuhyung Jo
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - So Dam Shin
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Chan-Sik Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Eun Jin Sohn
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Seon Gi Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseongdaero, Yuseong-gu, Daejeon 305-811, Republic of Korea
- *Jin Sook Kim:
| |
Collapse
|
35
|
Acute kidney injury: a conspiracy of Toll-like receptor 4 on endothelia, leukocytes, and tubules. Pediatr Nephrol 2012; 27:1847-54. [PMID: 22033798 PMCID: PMC3523189 DOI: 10.1007/s00467-011-2029-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/31/2011] [Accepted: 09/01/2011] [Indexed: 01/08/2023]
Abstract
Ischemic acute kidney injury (AKI) contributes to considerable morbidity and mortality in hospitalized patients and can contribute to rejection during kidney transplantation. Maladaptive immune responses can exacerbate injury, and targeting these responses holds promise as therapy for AKI. In the last decade, a number of molecules and receptors were identified in the innate immune response to ischemia-reperfusion injury. This review primarily focuses on one pathway that leads to maladaptive inflammation: toll-like receptor 4 (TLR4) and one of its ligands, high mobility group box protein 1 (HMGB1). The temporal-spatial roles and potential therapeutics targeting this particular receptor-ligand interaction are also explored.
Collapse
|
36
|
Arshad MI, Piquet-Pellorce C, Samson M. IL-33 and HMGB1 alarmins: sensors of cellular death and their involvement in liver pathology. Liver Int 2012; 32:1200-10. [PMID: 22530772 DOI: 10.1111/j.1478-3231.2012.02802.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 03/12/2012] [Indexed: 12/20/2022]
Abstract
'Alarmins' are a group of proteins or molecules that are released from cells during cellular demise to alert the host immune system. Two of them, Interleukin-33 (IL-33) and high-mobility group box-1 (HMGB1), share many similarities of cellular localization, functions and involvement in various inflammatory pathologies including hepatitis. The expressions of IL-33 and HMGB1, and their receptors ST2 and receptor for advanced glycation end products (RAGE), are substantially up-regulated during acute and chronic hepatitis. Recent data evidence a possible protective role of IL-33/ST2 axis during liver injury. A contrast in expression of IL-33 and HMGB1 alarmins were associated with type of hepatocellular death mediated by immune cells or hepato-toxic agents. The massive release of active form of IL-33 from hepatocytes may affect the recruitment and activation of its ST2-positive target immune cells in the liver to confer its alarmin functions. This review highlights the emerging roles of alarmin proteins in various liver pathologies, by focusing on classical HMGB1 and a newly discovered alarmin, the IL-33.
Collapse
Affiliation(s)
- Muhammad I Arshad
- Institut de Recherche Santé Environnement & Travail, Institut National de la Santé et de la Recherche Médicale (Inserm), U.1085, Rennes, France
| | | | | |
Collapse
|
37
|
The Role of Glycyrrhizin, an Inhibitor of HMGB1 Protein, in Anticancer Therapy. Arch Immunol Ther Exp (Warsz) 2012; 60:391-9. [DOI: 10.1007/s00005-012-0183-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 05/11/2012] [Indexed: 12/22/2022]
|
38
|
Ohnishi M, Katsuki H, Fukutomi C, Takahashi M, Motomura M, Fukunaga M, Matsuoka Y, Isohama Y, Izumi Y, Kume T, Inoue A, Akaike A. HMGB1 inhibitor glycyrrhizin attenuates intracerebral hemorrhage-induced injury in rats. Neuropharmacology 2011; 61:975-80. [DOI: 10.1016/j.neuropharm.2011.06.026] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Revised: 06/24/2011] [Accepted: 06/26/2011] [Indexed: 02/05/2023]
|
39
|
Early interleukin 6 production by leukocytes during ischemic acute kidney injury is regulated by TLR4. Kidney Int 2011; 80:504-15. [PMID: 21633411 DOI: 10.1038/ki.2011.140] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Although leukocytes infiltrate the kidney during ischemic acute kidney injury (AKI) and release interleukin 6 (IL6), their mechanism of activation is unknown. Here, we tested whether Toll-like receptor 4 (TLR4) on leukocytes mediated this activation by interacting with high-mobility group protein B1 (HMGB1) released by renal cells as a consequence of ischemic kidney injury. We constructed radiation-induced bone marrow chimeras using C3H/HeJ and C57BL/10ScNJ strains of TLR4 (-/-) mice and their respective TLR4 (+/+) wild-type counterparts and studied them at 4 h after an ischemic insult. Leukocytes adopted from TLR4 (+/+) mice infiltrated the kidneys of TLR4 (-/-) mice, and TLR4 (-/-) leukocytes infiltrated the kidneys of TLR4 (+/+) mice but caused little functional renal impairment in each case. Maximal ischemic AKI required both radiosensitive leukocytes and radioresistant renal parenchymal and endothelial cells from TLR4 (+/+) mice. Only TLR4 (+/+) leukocytes produced IL6 in vivo and in response to HMGB1 in vitro. Thus, following infiltration of the injured kidney, leukocytes produce IL6 when their TLR4 receptors interact with HMGB1 released by injured renal cells. This underscores the importance of TLR4 in the pathogenesis of ischemic AKI.
Collapse
|
40
|
Zampell JC, Yan A, Avraham T, Andrade V, Malliaris S, Aschen S, Rockson SG, Mehrara BJ. Temporal and spatial patterns of endogenous danger signal expression after wound healing and in response to lymphedema. Am J Physiol Cell Physiol 2011; 300:C1107-21. [PMID: 21248077 DOI: 10.1152/ajpcell.00378.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While acute tissue injury potently induces endogenous danger signal expression, the role of these molecules in chronic wound healing and lymphedema is undefined. The purpose of this study was to determine the spatial and temporal expression patterns of the endogenous danger signals high-mobility group box 1 (HMGB1) and heat shock protein (HSP)70 during wound healing and chronic lymphatic fluid stasis. In a surgical mouse tail model of tissue injury and lymphedema, HMGB1 and HSP70 expression occurred along a spatial gradient relative to the site of injury, with peak expression at the wound and greater than twofold reduced expression within 5 mm (P < 0.05). Expression primarily occurred in cells native to injured tissue. In particular, HMGB1 was highly expressed by lymphatic endothelial cells (>40% positivity; twofold increase in chronic inflammation, P < 0.001). We found similar findings using a peritoneal inflammation model. Interestingly, upregulation of HMGB1 (2.2-fold), HSP70 (1.4-fold), and nuclear factor (NF)-κβ activation persisted at least 6 wk postoperatively only in lymphedematous tissues. Similarly, we found upregulation of endogenous danger signals in soft tissue of the arm after axillary lymphadenectomy in a mouse model and in matched biopsy samples obtained from patients with secondary lymphedema comparing normal to lymphedematous arms (2.4-fold increased HMGB1, 1.9-fold increased HSP70; P < 0.01). Finally, HMGB1 blockade significantly reduced inflammatory lymphangiogenesis within inflamed draining lymph nodes (35% reduction, P < 0.01). In conclusion, HMGB1 and HSP70 are expressed along spatial gradients and upregulated in chronic lymphatic fluid stasis. Furthermore, acute expression of endogenous danger signals may play a role in inflammatory lymphangiogenesis.
Collapse
Affiliation(s)
- Jamie C Zampell
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ledford JG, Lo B, Kislan MM, Thomas JM, Evans K, Cain DW, Kraft M, Williams KL, Wright JR. Surfactant protein-A inhibits mycoplasma-induced dendritic cell maturation through regulation of HMGB-1 cytokine activity. THE JOURNAL OF IMMUNOLOGY 2010; 185:3884-94. [PMID: 20810986 DOI: 10.4049/jimmunol.1000387] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During pulmonary infections, a careful balance between activation of protective host defense mechanisms and potentially injurious inflammatory processes must be maintained. Surfactant protein A (SP-A) is an immune modulator that increases pathogen uptake and clearance by phagocytes while minimizing lung inflammation by limiting dendritic cell (DC) and T cell activation. Recent publications have shown that SP-A binds to and is bacteriostatic for Mycoplasma pneumoniae in vitro. In vivo, SP-A aids in maintenance of airway homeostasis during M. pneumoniae pulmonary infection by preventing an overzealous proinflammatory response mediated by TNF-α. Although SP-A was shown to inhibit maturation of DCs in vitro, the consequence of DC/SP-A interactions in vivo has not been elucidated. In this article, we show that the absence of SP-A during M. pneumoniae infection leads to increased numbers of mature DCs in the lung and draining lymph nodes during the acute phase of infection and, consequently, increased numbers of activated T and B cells during the course of infection. The findings that glycyrrhizin, a specific inhibitor of extracellular high-mobility group box-1 (HMGB-1) abrogated this effect and that SP-A inhibits HMGB-1 release from immune cells suggest that SP-A inhibits M. pneumoniae-induced DC maturation by regulating HMGB-1 cytokine activity.
Collapse
Affiliation(s)
- Julie G Ledford
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Weinberg EO. ST2 protein in heart disease: from discovery to mechanisms and prognostic value. Biomark Med 2009; 3:495-511. [DOI: 10.2217/bmm.09.56] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Biomarkers aid in diagnosis by providing important information for the clinical assessment of patients that is not achieved by other means. This article focuses on the use of soluble ST2 as a biomarker in cardiovascular disease. Soluble ST2 is a secreted receptor belonging to the IL-1 receptor family that regulates inflammation and immunity. Recent studies have shown that soluble ST2 is a decoy receptor that disrupts the interaction of IL-33 with the full-length ST2L receptor. The rapidly evolving and expanding literature on ST2 and its ligand reveal emerging roles for this system in the regulation of inflammation in a variety of diseases. Elevated ST2 levels have been detected in cardiovascular diseases and levels provide useful prognostic information in many, but not all, of these diseases, which will be discussed here. Additional studies demonstrating elevated soluble ST2 levels in other human diseases will also be discussed.
Collapse
Affiliation(s)
- Ellen O Weinberg
- Boston Medical Center, EBRC Room 704, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|