1
|
Mizrachi M, Diamond B. Impact of microglia isolation and culture methodology on transcriptional profile and function. J Neuroinflammation 2024; 21:87. [PMID: 38589917 PMCID: PMC11000335 DOI: 10.1186/s12974-024-03076-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND Microglial isolation and culturing methods continue to be explored to maximize cellular yield, purity, responsiveness to stimulation and similarity to in vivo microglia. This study aims to evaluate five different microglia isolation methods-three variants of microglia isolation from neonatal mice and two variants of microglia isolation from adult mice-on transcriptional profile and response to HMGB1. METHODS Microglia from neonatal mice, age 0-3 days (P0-P3) were isolated from mixed glial cultures (MGC). We included three variations of this protocol that differed by use of GM-CSF in culture (No GM-CSF or 500 pg/mL GM-CSF), and days of culture in MGC before microglial separation (10 or 21). Protocols for studying microglia from adult mice age 6-8 weeks included isolation by adherence properties followed by 7 days of culture with 100 ng/mL GM-CSF and 100 ng/mL M-CSF (Vijaya et al. in Front Cell Neurosci 17:1082180, 2023), or acute isolation using CD11b beads (Bordt et al. in STAR Protoc 1:100035, 2020. https://doi.org/10.1016/j.xpro.2020.100035 ). Purity, yield, and RNA quality of the isolated microglia were assessed by flow cytometry, hemocytometer counting, and Bioanalyzer, respectively. Microglial responsiveness to an inflammatory stimulus, HMGB1, was evaluated by measuring TNFα, IL1β, and IFNβ concentration in supernatant by ELISA and assessing gene expression patterns using bulk mRNA sequencing. RESULTS All five methods demonstrated greater than 90% purity. Microglia from all cultures increased transcription and secretion of TNFα, IL1β, and IFNβ in response to HMGB1. RNA sequencing showed a larger number of differentially expressed genes in response to HMGB1 treatment in microglia cultured from neonates than from adult mice, with sparse changes among the three MGC culturing conditions. Additionally, cultured microglia derived from adult and microglia derived from MGCs from neonates display transcriptional signatures corresponding to an earlier developmental stage. CONCLUSION These findings suggest that while all methods provided high purity, the choice of protocol may significantly influence yield, RNA quality, baseline transcriptional profile and response to stimulation. This comparative study provides valuable insights to inform the choice of microglial isolation and culture method.
Collapse
Affiliation(s)
- Mark Mizrachi
- Feinstein Institutes of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA
| | - Betty Diamond
- Feinstein Institutes of Molecular Medicine, Feinstein Institutes for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, 500 Hofstra Blvd, Hempstead, NY, 11549, USA.
| |
Collapse
|
2
|
Presto P, Ji G, Ponomareva O, Ponomarev I, Neugebauer V. Hmgb1 Silencing in the Amygdala Inhibits Pain-Related Behaviors in a Rat Model of Neuropathic Pain. Int J Mol Sci 2023; 24:11944. [PMID: 37569320 PMCID: PMC10418916 DOI: 10.3390/ijms241511944] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/16/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
Chronic pain presents a therapeutic challenge due to the highly complex interplay of sensory, emotional-affective and cognitive factors. The mechanisms of the transition from acute to chronic pain are not well understood. We hypothesized that neuroimmune mechanisms in the amygdala, a brain region involved in the emotional-affective component of pain and pain modulation, play an important role through high motility group box 1 (Hmgb1), a pro-inflammatory molecule that has been linked to neuroimmune signaling in spinal nociception. Transcriptomic analysis revealed an upregulation of Hmgb1 mRNA in the right but not left central nucleus of the amygdala (CeA) at the chronic stage of a spinal nerve ligation (SNL) rat model of neuropathic pain. Hmgb1 silencing with a stereotaxic injection of siRNA for Hmgb1 into the right CeA of adult male and female rats 1 week after (post-treatment), but not 2 weeks before (pre-treatment) SNL induction decreased mechanical hypersensitivity and emotional-affective responses, but not anxiety-like behaviors, measured 4 weeks after SNL. Immunohistochemical data suggest that neurons are a major source of Hmgb1 in the CeA. Therefore, Hmgb1 in the amygdala may contribute to the transition from acute to chronic neuropathic pain, and the inhibition of Hmgb1 at a subacute time point can mitigate neuropathic pain.
Collapse
Affiliation(s)
- Peyton Presto
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Guangchen Ji
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Olga Ponomareva
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Igor Ponomarev
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Volker Neugebauer
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Center of Excellence for Translational Neuroscience and Therapeutics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
3
|
Banks WA, Hansen KM, Erickson MA, Crews FT. High-mobility group box 1 (HMGB1) crosses the BBB bidirectionally. Brain Behav Immun 2023; 111:386-394. [PMID: 37146655 DOI: 10.1016/j.bbi.2023.04.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/07/2023] Open
Abstract
High-mobility group box 1 (HMGB1) is a ubiquitous protein that regulates transcription in the nucleus, and is an endogenous damage-associated molecular pattern molecule that activates the innate immune system. HMGB1 activates the TLR4 and RAGE recepto, inducing downstream signals reminiscent of cytokines that have been found to cross the blood-brain barrier (BBB). Blood HMGB1 increases in stroke, sepsis, senescence, alcohol binge drinking and other conditions. Here, we examined the ability of HMGB1 radioactively labeled with iodine (I-HMGB1) to cross the BBB. We found that I-HMGB1 readily entered into mouse brain from the circulation with a unidirectional influx rate of 0.654 μl/g-min. All brain regions tested took up I-HMGB1; uptake was greatest by the olfactory bulb and least in the striatum. Transport was not reliably inhibited by unlabeled HMGB1 nor by inhibitors of TLR4, TLR2, RAGE, or CXCR4. Uptake was enhanced by co-injection of wheatgerm agglutinin, suggestive of involvement of absorptive transcytosis as a mechanism of transport. Induction of inflammation/neuroinflammation with lipopolysaccharide is known to increase blood HMGB1; we report here that brain transport is also increased by LPS-induced inflammation. Finally, we found that I-HMGB1 was also transported in the brain-to-blood direction, with both unlabeled HMGB1 or lipopolysaccharide increasing the transport rate. These results show that HMGB1 can bidirectionally cross the BBB and that those transport rates are enhanced by inflammation. Such transport provides a mechanism by which HMGB1 levels would impact neuroimmune signaling in both the brain and periphery.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State.
| | - Kim M Hansen
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Michelle A Erickson
- Geriatric Research Educational and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, US State; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA, US State
| | - Fulton T Crews
- Department of Pharmacology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, US State
| |
Collapse
|
4
|
Rızvanoglu İH, Sakarya B, Benlier N, Kökçü F. HMGB-1 Levels in Painful Knee Arthroplasty: Is it Possible to Distingue Periprosthetic Joint Infection and Aseptic Loosening? Indian J Orthop 2023; 57:1023-1031. [PMID: 37384002 PMCID: PMC10293519 DOI: 10.1007/s43465-023-00903-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/27/2023] [Indexed: 06/30/2023]
Abstract
Introduction We investigated the efficiency of high mobility group box-1 protein (HMGB-1) in differentiation of asymptomatic knee prosthesis, and periprosthetic joint infection and aseptic loosening causing painful knee prosthesis. Materials and Methods The data of patients who consulted our clinic for checking after total knee arthroplasty surgery were recorded prospectively. Blood levels of CRP, ESR, WBC, and HMGB-1 were recorded. Patients whose examination and routine tests were within normal limits comprised group I, asymptomatic total knee arthroplasty (ATKA). Painful patients with abnormal test results underwent three phase bone scintigraphy for further investigation Patients with periprosthetic joint infection (PJI) and aseptic loosening (AL) according to scintigraphy comprised group II and group III, respectively. The mean values of HMGB-1 and cut-off values according to the groups and their correlations with other inflammatory parameters were determined. Results Seventy-three patients were included in the study. Significant differences were observed in three groups, in terms of CRP, ESR, WBC, and HMGB-1. The cut-off value of HMGB-1 was determined as 15.16 ng/ml between ATKA and PJI, 16.92 ng/ml between ATKA and AL, and 27.87 ng/ml between PJI and AL, respectively. Accordingly, the sensitivity, and specificity of HMGB-1 in differentiation of ATKA and PJI were 91%, 88%, and in differentiation of ATKA and AL were 91%, 96%, and in differentiation of PJI and AL were 81%, 73%, respectively. Conclusion HMGB-1 may be utilized as an additional blood test in the differential diagnosis of problematic knee prosthesis patients.
Collapse
Affiliation(s)
- İbrahim Halil Rızvanoglu
- Department of Orthopedics and Traumatology, NCR International Hospital, Mücahitler Neighborhood, No: 56. Şehitkamil, 27090 Gaziantep, Turkey
| | - Bülent Sakarya
- Department of Orthopedics and Traumatology, Mersin Toros Government Hospital, Cemalpaşa Neighborhood. 5314 Street, Akdeniz, Mersin, Turkey
| | - Necla Benlier
- Department of Medical Pharmacology, Sanko University School of Medicine, Ali Fuat Cebesoy Boulevard. No: 45, Şehitkamil, Gaziantep, Turkey
| | - Füsun Kökçü
- Department of Nuclear Medicine, Medical Park Gaziantep Hospital, Mücahitler Neighborhood, 52063 St., Şehitkamil, 27584 Gaziantep, Turkey
| |
Collapse
|
5
|
Electroacupuncture Alleviates Neuroinflammation by Inhibiting the HMGB1 Signaling Pathway in Rats with Sepsis-Associated Encephalopathy. Brain Sci 2022; 12:brainsci12121732. [PMID: 36552192 PMCID: PMC9776077 DOI: 10.3390/brainsci12121732] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/29/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Sepsis-Associated Encephalopathy (SAE) is common in sepsis patients, with high mortality rates. It is believed that neuroinflammation is an important mechanism involved in SAE. High mobility group box 1 protein (HMGB1), as a late pro-inflammatory factor, is significantly increased during sepsis in different brain regions, including the hippocampus. HMGB1 causes neuroinflammation and cognitive impairment through direct binding to advanced glycation end products (RAGE) and Toll-like receptor 4 (TLR4). Electroacupuncture (EA) at Baihui (GV20) and Zusanli (ST36) is beneficial for neurological diseases and experimental sepsis. Our study used EA to treat SAE induced by lipopolysaccharide (LPS) in male Sprague-Dawley rats. The Y maze test was performed to assess working memory. Immunofluorescence (IF) and Western blotting (WB) were used to determine neuroinflammation and the HMGB1 signaling pathway. Results showed that EA could improve working memory impairment in rats with SAE. EA alleviated neuroinflammation by downregulating the hippocampus's HMGB1/TLR4 and HMGB1/RAGE signaling, reducing the levels of pro-inflammatory factors, and relieving microglial and astrocyte activation. However, EA did not affect the tight junctions' expression of the blood-brain barrier (BBB) in the hippocampus.
Collapse
|
6
|
Yang J, Ran M, Li H, Lin Y, Ma K, Yang Y, Fu X, Yang S. New insight into neurological degeneration: Inflammatory cytokines and blood–brain barrier. Front Mol Neurosci 2022; 15:1013933. [PMID: 36353359 PMCID: PMC9637688 DOI: 10.3389/fnmol.2022.1013933] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/20/2022] [Indexed: 11/30/2022] Open
Abstract
Neurological degeneration after neuroinflammation, such as that resulting from Alzheimer’s disease (AD), stroke, multiple sclerosis (MS), and post-traumatic brain injury (TBI), is typically associated with high mortality and morbidity and with permanent cognitive dysfunction, which places a heavy economic burden on families and society. Diagnosing and curing these diseases in their early stages remains a challenge for clinical investigation and treatment. Recent insight into the onset and progression of these diseases highlights the permeability of the blood–brain barrier (BBB). The primary factor that influences BBB structure and function is inflammation, especially the main cytokines including IL-1β, TNFα, and IL-6, the mechanism on the disruption of which are critical component of the aforementioned diseases. Surprisingly, the main cytokines from systematic inflammation can also induce as much worse as from neurological diseases or injuries do. In this review, we will therefore discuss the physiological structure of BBB, the main cytokines including IL-1β, TNFα, IL-6, and their mechanism on the disruption of BBB and recent research about the main cytokines from systematic inflammation inducing the disruption of BBB and cognitive impairment, and we will eventually discuss the need to prevent the disruption of BBB.
Collapse
Affiliation(s)
- Jie Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Mingzi Ran
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Anaesthesiology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Hongyu Li
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Ye Lin
- Department of Neurology, The First Medical Centre, PLA General Hospital, Beijing, China
| | - Kui Ma
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
| | - Yuguang Yang
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
| | - Xiaobing Fu
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Xiaobing Fu,
| | - Siming Yang
- Research Centre for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Department, PLA General Hospital, PLA Medical College, Beijing, China
- Department of Dermatology, 4th Medical Centre, PLA General Hospital, Beijing, China
- *Correspondence: Siming Yang,
| |
Collapse
|
7
|
Melatonin attenuates bone cancer pain via the SIRT1/HMGB1 pathway. Neuropharmacology 2022; 220:109254. [PMID: 36122662 DOI: 10.1016/j.neuropharm.2022.109254] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/08/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022]
Abstract
Bone cancer pain (BCP), which seriously affects the quality of life of patients, remains a clinically challenging problem. Hence, there is an urgent need to investigate new mechanisms and develop new therapeutics to relieve BCP. In the present study, we investigated the analgesic effect of melatonin on BCP and the underlying mechanisms. Male C57BL/6 mice were used to establish BCP models. We found that the levels of sirtuin 1 (SIRT1) and nucleus-high mobility group box-1 (HMGB1) were decreased, whilst the levels of HMGB1, cytoplasm-HMGB1 and inflammatory cytokines (TNF-α, IL-6, IL-1β) were increased in the spinal cord of BCP mice on days 7, 14 and 21 after implantation compared with the levels in sham mice. Intrathecal administration of melatonin dose-dependently increased values of PWMT and TWL compared with the BCP group. However, intrathecal administration of EX527 (a selective SIRT1 antagonist) reversed the analgesic effect of melatonin. Moreover, mice in the melatonin group exhibited an increase in SIRT1 and nucleus-HMGB1, whilst there was a decrease in HMGB1, cytoplasm-HMGB1, rage, acetyl-HMGB1 and inflammatory cytokines compared with those in BCP mice. EX527 also reversed these changes. Furthermore, SIRT1 physically interacted with HMGB1 in the BCP mice. In conclusion, intrathecal administration of melatonin attenuates BCP through SIRT1-dependent inhibition of HMGB1 translocation and inflammatory cytokines. Melatonin may be a promising drug for the clinical treatment of BCP.
Collapse
|
8
|
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: 14] [Impact Index Per Article: 4.7] [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
|
9
|
Rouillard ME, Hu J, Sutter PA, Kim HW, Huang JK, Crocker SJ. The Cellular Senescence Factor Extracellular HMGB1 Directly Inhibits Oligodendrocyte Progenitor Cell Differentiation and Impairs CNS Remyelination. Front Cell Neurosci 2022; 16:833186. [PMID: 35573828 PMCID: PMC9095917 DOI: 10.3389/fncel.2022.833186] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
HMGB1 is a highly conserved, ubiquitous protein in eukaryotic cells. HMGB1 is normally localized to the nucleus, where it acts as a chromatin associated non-histone binding protein. In contrast, extracellular HMGB1 is an alarmin released by stressed cells to act as a danger associated molecular pattern (DAMP). We have recently determined that progenitor cells from multiple sclerosis patients exhibit a cellular senescent phenotype and release extracellular HMGB1 which directly impaired the maturation of oligodendrocyte progenitor cells (OPCs) to myelinating oligodendrocytes (OLs). Herein, we report that administration of recombinant HMGB1 into the spinal cord at the time of lysolecithin administration resulted in arrest of OPC differentiation in vivo, and a profound impairment of remyelination. To define the receptor by which extracellular HMGB1 mediates its inhibitory influence on OPCs to impair OL differentiation, we tested selective inhibitors against the four primary receptors known to mediate the effects of HMGB1, the toll-like receptors (TLRs)-2, -4, -9 or the receptor for advanced glycation end-products (RAGE). We found that inhibition of neither TLR9 nor RAGE increased OL differentiation in the presence of HMGB1, while inhibition of TLR4 resulted in partial restoration of OL differentiation and inhibiting TLR2 fully restored differentiation of OLs in the presence of HMGB1. Analysis of transcriptomic data (RNAseq) from OPCs identified an overrepresentation of NFκB regulated genes in OPCs when in the presence of HMGB1. We found that application of HMGB1 to OPCs in culture resulted in a rapid and concentration dependent shift in NFκB nuclear translocation which was also attenuated with coincident TLR2 inhibition. These data provide new information on how extracellular HMGB1 directly affects the differentiation potential of OPCs. Recent and past evidence for elevated HMGB1 released from senescent progenitor cells within demyelinated lesions in the MS brain suggests that a greater understanding of how this molecule acts on OPCs may unfetter the endogenous remyelination potential in MS.
Collapse
Affiliation(s)
- Megan E. Rouillard
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Jingwen Hu
- Department of Biology and Center for Cell Reprogramming, Georgetown University, Washington, DC, United States
| | - Pearl A. Sutter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Hee Won Kim
- Department of Biology and Center for Cell Reprogramming, Georgetown University, Washington, DC, United States
| | - Jeffrey K. Huang
- Department of Biology and Center for Cell Reprogramming, Georgetown University, Washington, DC, United States
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
10
|
Sensory Neuron TLR4 mediates the development of nerve-injury induced mechanical hypersensitivity in female mice. Brain Behav Immun 2021; 97:42-60. [PMID: 34174335 PMCID: PMC8453057 DOI: 10.1016/j.bbi.2021.06.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 02/07/2023] Open
Abstract
Recent studies have brought to light the necessity to discern sex-specific differences in various pain states and different cell-types that mediate these differences. These studies have uncovered the role of neuroimmune interactions to mediate pain states in a sex-specific fashion. While investigating immune function in pain development, we discovered that females utilize immune components of sensory neurons to mediate neuropathic pain development. We utilized two novel transgenic mouse models that eitherrestore expression of toll-like receptor (TLR) 4 inNav1.8 nociceptors on a TLR4-null background (TLR4LoxTB) or remove TLR4 specifically from Nav1.8 nociceptors (TLR4fl/fl). After spared nerve injury (SNI), a model of neuropathic injury, we observed a robust female-specific onset of mechanical hypersensitivity in our transgenic animals. Female Nav1.8-TLR4fl/fl knockout animals were less mechanically sensitive than cre-negative TLR4fl/fl littermates. Conversely, female Nav1.8-TLR4LoxTB reactivated animals were as mechanically sensitive as their wild-type counterparts. These sex and cell-specific effects were not recapitulated in male animals of either strain. Additionally, we find the danger associated molecular pattern, high mobility group box-1 (HGMB1), a potent TLR4 agonist, localization and ATF3 expression in females is dependent on TLR4 expression in dorsal root ganglia (DRG) populations following SNI. These experiments provide novel evidence toward sensory neuron specific modulation of pain in a sex-dependent manner.
Collapse
|
11
|
Lee JH, Kim HS, Lee D, Yun T, Koo Y, Chae Y, Kang JH, Kang BT, Yang MP, Kim H. Clinical signs, duodenal histopathological grades, and serum high-mobility group box 1 concentrations in dogs with inflammatory bowel disease. J Vet Intern Med 2021; 35:2205-2214. [PMID: 34480505 PMCID: PMC8478061 DOI: 10.1111/jvim.16258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/25/2022] Open
Abstract
Background Inflammatory bowel disease (IBD) commonly occurs in dogs, but there is lack of information about potential biomarkers of clinical and histopathologic severity. Objective To examine the role of serum C‐reactive protein (CRP) and high‐mobility group box 1 (HMGB1) concentrations in dogs with IBD. Animals Seventeen dogs with IBD and 25 healthy dogs. Methods In this prospective study, duodenal histopathologic severity was graded, and the clinical severity of IBD was assessed by the canine IBD assessment index (CIBDAI) score in dogs with IBD. Serum CRP and HMGB1 concentrations were compared between IBD and healthy dogs and analyzed according to histopathologic grade in dogs with IBD. The correlations between serum CRP and HMGB1 concentrations and the CIBDAI score were evaluated. Results Dogs with IBD had higher serum CRP (median [range] = 20.39 [1.53‐67.69] μg/mL vs 2.31 [0.17‐11.49] μg/mL; P < .001) and HMGB1 concentrations (0.44 [0.07‐1.58] ng/mL vs 0.05 [0.01‐0.25] ng/mL; P < .001) than healthy dogs. The serum HMGB1 concentration was higher in IBD dogs with a moderate to severe histopathologic grade (0.51 [0.30‐1.58] ng/mL, P = .03) than in those with a mild histopathologic grade (0.17 [0.07‐0.75] ng/mL). Serum CRP concentrations and CIBDAI score were positively correlated in dogs with IBD (rs = .49, P = .05). Conclusions and Clinical Importance Serum HMGB1 could be a potential biomarker for diagnosing IBD and might be indicative of histopathologic severity in dogs, whereas serum CRP might be an indicator of clinical severity.
Collapse
Affiliation(s)
- Jong-Hwan Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hong-Suk Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Dohee Lee
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Taesik Yun
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Yoonhoi Koo
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Yeon Chae
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ji-Houn Kang
- Western Animal Medical Center, Seoul, Republic of Korea
| | - Byeong-Teck Kang
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Mhan-Pyo Yang
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Hakhyun Kim
- Laboratory of Veterinary Internal Medicine, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| |
Collapse
|
12
|
Circulating HMGB1 is elevated in veterans with Gulf War Illness and triggers the persistent pro-inflammatory microglia phenotype in male C57Bl/6J mice. Transl Psychiatry 2021; 11:390. [PMID: 34253711 PMCID: PMC8275600 DOI: 10.1038/s41398-021-01517-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/16/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Gulf War Illness (GWI) is a chronic, multi-symptom peripheral and CNS condition with persistent microglial dysregulation, but the mechanisms driving the continuous neuroimmune pathology are poorly understood. The alarmin HMGB1 is an autocrine and paracrine pro-inflammatory signal, but the role of circulating HMGB1 in persistent neuroinflammation and GWI remains largely unknown. Using the LPS model of the persistent microglial pro-inflammatory response, male C57Bl/6J mice injected with LPS (5 mg/kg IP) exhibited persistent changes in microglia morphology and elevated pro-inflammatory markers in the hippocampus, cortex, and midbrain 7 days after LPS injection, while the peripheral immune response had resolved. Ex vivo serum analysis revealed an augmented pro-inflammatory response to LPS when microglia cells were cultured with the 7-day LPS serum, indicating the presence of bioactive circulating factors that prime the microglial pro-inflammatory response. Elevated circulating HMGB1 levels were identified in the mouse serum 7 days after LPS administration and in the serum of veterans with GWI. Tail vein injection of rHMGB1 in male C57Bl/6 J mice elevated TNFα mRNA levels in the liver, hippocampus, and cortex, demonstrating HMGB1-induced peripheral and CNS effects. Microglia isolated at 7 days after LPS injection revealed a unique transcriptional profile of 17 genes when compared to the acute 3 H LPS response, 6 of which were also upregulated in the midbrain by rHMGB1, highlighting a distinct signature of the persistent pro-inflammatory microglia phenotype. These findings indicate that circulating HMGB1 is elevated in GWI, regulates the microglial neuroimmune response, and drives chronic neuroinflammation that persists long after the initial instigating peripheral stimulus.
Collapse
|
13
|
Peek V, Harden LM, Damm J, Aslani F, Leisengang S, Roth J, Gerstberger R, Meurer M, von Köckritz-Blickwede M, Schulz S, Spengler B, Rummel C. LPS Primes Brain Responsiveness to High Mobility Group Box-1 Protein. Pharmaceuticals (Basel) 2021; 14:ph14060558. [PMID: 34208101 PMCID: PMC8230749 DOI: 10.3390/ph14060558] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/30/2022] Open
Abstract
High mobility group box (HMGB)1 action contributes to late phases of sepsis, but the effects of increased endogenous plasma HMGB1 levels on brain cells during inflammation are unclear. Here, we aimed to further investigate the role of HMGB1 in the brain during septic-like lipopolysaccharide-induced inflammation in rats (LPS, 10 mg/kg, i.p.). HMGB-1 mRNA expression and release were measured in the periphery/brain by RT-PCR, immunohistochemistry and ELISA. In vitro experiments with disulfide-HMGB1 in primary neuro-glial cell cultures of the area postrema (AP), a circumventricular organ with a leaky blood–brain barrier and direct access to circulating mediators like HMGB1 and LPS, were performed to determine the direct influence of HMGB1 on this pivotal brain structure for immune-to-brain communication. Indeed, HMGB1 plasma levels stayed elevated after LPS injection. Immunohistochemistry of brains and AP cultures confirmed LPS-stimulated cytoplasmatic translocation of HMGB1 indicative of local HMGB1 release. Moreover, disulfide-HMGB1 stimulation induced nuclear factor (NF)-κB activation and a significant release of interleukin-6, but not tumor necrosis factor α, into AP culture supernatants. However, only a few AP cells directly responded to HMGB1 with increased intracellular calcium concentration. Interestingly, priming with LPS induced a seven-fold higher percentage of responsive cells to HMGB1. We conclude that, as a humoral and local mediator, HMGB1 enhances brain inflammatory responses, after LPS priming, linked to sustained sepsis symptoms.
Collapse
Affiliation(s)
- Verena Peek
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
| | - Lois M. Harden
- Brain Function Research Group, School of Physiology, Faculty of Health Sciences, University of Witwatersrand, Johannesburg 2193, South Africa;
| | - Jelena Damm
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
| | - Ferial Aslani
- Institute of Anatomy and Cell Biology of the Medical Faculty, Justus Liebig University, 35392 Giessen, Germany;
| | - Stephan Leisengang
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
| | - Joachim Roth
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
| | - Rüdiger Gerstberger
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
| | - Marita Meurer
- Department of Biochemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (M.M.); (M.v.K.-B.)
| | - Maren von Köckritz-Blickwede
- Department of Biochemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany and Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hannover, 30559 Hannover, Germany; (M.M.); (M.v.K.-B.)
| | - Sabine Schulz
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (S.S.); (B.S.)
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (S.S.); (B.S.)
| | - Christoph Rummel
- Institute of Veterinary Physiology and Biochemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (V.P.); (J.D.); (S.L.); (J.R.); (R.G.)
- Correspondence:
| |
Collapse
|
14
|
He W, Xu F, Chen L, Huang W, Jiang L, Tang F, Yan W, Zhong S, Shen C, Huang H, Lv J, Wu X, Zeng S, Li M, Zhang M. Association of High-Mobility Group Box-1 with Inflammationrelated Cytokines in the Aqueous Humor with Acute Primary Angle-Closure Eyes. Curr Mol Med 2021; 21:237-245. [PMID: 32282301 DOI: 10.2174/1566524020666200413113107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/16/2020] [Accepted: 03/24/2020] [Indexed: 11/22/2022]
Abstract
AIM The aim of this study was to measure the levels of High-mobility group box-1 (HMGB1) and inflammation-related cytokines in the aqueous humor of patients with acute primary angle-closure glaucoma (APAG) and age-related cataract eyes (ARC). METHODS Aqueous humor samples were obtained from 59 eyes of 59 Chinese subjects (APAG, 32 eyes; and ARC, 27eyes). The multiplex bead immunoassay technique was used to measure the levels of HMGB1 and IL-8, IL-6, G-CSF, MCP-3, VEGF, sVEGFR- 1, sVEFGR-2, TNF-α, PDGF, and IL-10 in aqueous. The data of Patients' demographics and preoperative intraocular pressure (IOP) were also collected for detailed analysis. RESULTS The APAG group showed significantly elevated concentrations of HMGB1, IL- 8, IL-6, G-CSF, VEGF, sVEGFR-1, and TNF-α than those in the ARC group. Aqueous HMGB1 level correlated significantly with IOP, IL-8, IL-6, G-CSF and sVEGFR-1 levels but not with age, TNF-α, or VEGF levels. CONCLUSION The aqueous level of HMGB1 is elevated in APAG and associated with aqueous level of inflammation-related cytokines, suggesting an association between elevated levels of HMGB1, APAC and certain inflammatory modulators which, of course, should lead to further investigations in order to demonstrate the cause and effect.
Collapse
Affiliation(s)
- Wenjing He
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Fan Xu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Lifei Chen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Wei Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Li Jiang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Fen Tang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Wenya Yan
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Shan Zhong
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Chaolan Shen
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Hui Huang
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Jian Lv
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Xiaonian Wu
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Siming Zeng
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Min Li
- Department of Ophthalmology, People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, Guangxi, China
| | - Mingyuan Zhang
- Laboratory Animal Center, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
15
|
Kwilasz AJ, Green Fulgham SM, Duran-Malle JC, Schrama AEW, Mitten EH, Todd LS, Patel HP, Larson TA, Clements MA, Harris KM, Litwiler ST, Harvey LO, Maier SF, Chavez RA, Rice KC, Van Dam AM, Watkins LR. Toll-like receptor 2 and 4 antagonism for the treatment of experimental autoimmune encephalomyelitis (EAE)-related pain. Brain Behav Immun 2021; 93:80-95. [PMID: 33358978 PMCID: PMC8475740 DOI: 10.1016/j.bbi.2020.12.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/26/2022] Open
Abstract
Neuropathic pain is a major symptom of multiple sclerosis (MS) with up to 92% of patients reporting bodily pain, and 85% reporting pain severe enough to cause functional disability. None of the available therapeutics target MS pain. Toll-like receptors 2 and 4 (TLR2/TLR4) have emerged as targets for treating a wide array of autoimmune disorders, including MS, as well as having demonstrated success at suppressing pain in diverse animal models. The current series of studies tested systemic TLR2/TLR4 antagonists in males and females in a low-dose Myelin oligodendrocyte glycoprotein (MOG) experimental autoimmune encephalomyelitis (EAE) model, with reduced motor dysfunction to allow unconfounded testing of allodynia through 50+ days post-MOG. The data demonstrated that blocking TLR2/TLR4 suppressed EAE-related pain, equally in males and females; upregulation of dorsal spinal cord proinflammatory gene expression for TLR2, TLR4, NLRP3, interleukin-1β, IkBα, TNF-α and interleukin-17; and upregulation of dorsal spinal cord expression of glial immunoreactivity markers. In support of these results, intrathecal interleukin-1 receptor antagonist reversed EAE-induced allodynia, both early and late after EAE induction. In contrast, blocking TLR2/TLR4 did not suppress EAE-induced motor disturbances induced by a higher MOG dose. These data suggest that blocking TLR2/TLR4 prevents the production of proinflammatory factors involved in low dose EAE pathology. Moreover, in this EAE model, TLR2/TLR4 antagonists were highly effective in reducing pain, whereas motor impairment, as seen in high dose MOG EAE, is not affected.
Collapse
Affiliation(s)
- Andrew J Kwilasz
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States.
| | - Suzanne M Green Fulgham
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Julissa Chante Duran-Malle
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Anouk E W Schrama
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Eric H Mitten
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Laurel S Todd
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Hardik P Patel
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Tracey A Larson
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Madison A Clements
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Kevin M Harris
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Scott T Litwiler
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Lewis O Harvey
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | - Steven F Maier
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| | | | - Kenner C Rice
- Drug Design and Synthesis Section, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, United States
| | - Anne-Marie Van Dam
- Department of Anatomy and Neuroscience, Amsterdam UMC, Vrije Universiteit, Amsterdam, Netherlands
| | - Linda R Watkins
- Department of Psychology and Neuroscience, University of Colorado, Boulder, CO, United States; The Center for Neuroscience, University of Colorado, Boulder, CO, United States
| |
Collapse
|
16
|
Agalave NM, Rudjito R, Farinotti AB, Khoonsari PE, Sandor K, Nomura Y, Szabo-Pardi TA, Urbina CM, Palada V, Price TJ, Erlandsson Harris H, Burton MD, Kultima K, Svensson CI. Sex-dependent role of microglia in disulfide high mobility group box 1 protein-mediated mechanical hypersensitivity. Pain 2021; 162:446-458. [PMID: 32773600 PMCID: PMC7808363 DOI: 10.1097/j.pain.0000000000002033] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/20/2020] [Accepted: 07/06/2020] [Indexed: 12/14/2022]
Abstract
ABSTRACT High mobility group box 1 protein (HMGB1) is increasingly regarded as an important player in the spinal regulation of chronic pain. Although it has been reported that HMGB1 induces spinal glial activation in a Toll-like receptor (TLR)4-dependent fashion, the aspect of sexual dimorphisms has not been thoroughly addressed. Here, we examined whether the action of TLR4-activating, partially reduced disulfide HMGB1 on microglia induces nociceptive behaviors in a sex-dependent manner. We found disulfide HMGB1 to equally increase microglial Iba1 immunoreactivity in lumbar spinal dorsal horn in male and female mice, but evoke higher cytokine and chemokine expression in primary microglial culture derived from males compared to females. Interestingly, TLR4 ablation in myeloid-derived cells, which include microglia, only protected male mice from developing HMGB1-induced mechanical hypersensitivity. Spinal administration of the glial inhibitor, minocycline, with disulfide HMGB1 also prevented pain-like behavior in male mice. To further explore sex difference, we examined the global spinal protein expression using liquid chromatography-mass spectrometry and found several antinociceptive and anti-inflammatory proteins to be upregulated in only male mice subjected to minocycline. One of the proteins elevated, alpha-1-antitrypsin, partially protected males but not females from developing HMGB1-induced pain. Targeting downstream proteins of alpha-1-antitrypsin failed to produce robust sex differences in pain-like behavior, suggesting that several proteins identified by liquid chromatography-mass spectrometry are required to modulate the effects. Taken together, the current study highlights the importance of mapping sex dimorphisms in pain mechanisms and point to processes potentially involved in the spinal antinociceptive effect of microglial inhibition in male mice.
Collapse
Affiliation(s)
- Nilesh M. Agalave
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Neuroscience, Neuroimmunology and Behavior Group, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Resti Rudjito
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alex Bersellini Farinotti
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Payam Emami Khoonsari
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yuki Nomura
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Thomas A. Szabo-Pardi
- Department of Neuroscience, Neuroimmunology and Behavior Group, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Carlos Morado Urbina
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Vinko Palada
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Theodore J. Price
- Department of Neuroscience, Pain Neurobiology Research Group, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States
| | | | - Michael D. Burton
- Department of Neuroscience, Neuroimmunology and Behavior Group, School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, United States
| | - Kim Kultima
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Camilla I. Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
17
|
Chibaatar E, Le K, Abdoulaye IA, Wu S, Guo Y. Melatonin Ameliorates Lipopolysaccharide-Induced Microglial Inflammation via Triggering SIRT1/HMGB1 Signaling Axis. J Mol Neurosci 2020; 71:691-701. [PMID: 32910356 DOI: 10.1007/s12031-020-01699-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022]
Abstract
Stroke is one of the highest incidence neurological disorder with great morbidity and mortality rate. The secondary neuroinflammation contributed by microglial activation is a consequential response observed in the pathogenesis of stroke. High-mobility group box 1, a non-histone nuclear protein, interacts with immune cells, such as microglia, and leads to a cascade amplification of the secondary neuroinflammatory responses, which are related to neuronal damage later. Melatonin is a neurohormone, well-known as its anti-oxidative and anti-inflammatory effects. However, until now, more findings are required for better understanding about anti-inflammatory effect of melatonin on HMGB1 and HMGB1-triggered pathway in LPS-induced microglial activation. Melatonin effect on the viability of BV2 microglial cells was measured by CCK-8 assay; mRNA levels of HMGB1 and other inflammatory cytokines were determined by quantitative real-time polymerase chain reaction assay or enzyme-linked immunosorbent assays; the protein expression levels of TLR4/MyD88/NF-κB and SIRT1 were detected by Western blot, and HMGB1 translocation and release from BV2 microglial cells were examined by immunofluorescence assay. The results of this study demonstrated that melatonin suppressed LPS-triggered BV2 microglial activation-mediated inflammation by inhibiting high expression and release of HMGB1 and moderating the activation of subsequent TLR4/MyD88/NF-κB signaling pathway, which was activated by SIRT1 elevation. Furthermore, LPS-induced expression of pro-inflammatory cytokines (i.e., TNF-α, IL-6, and IL-1β) was notably reversed by melatonin pre-treatment. In summary, our findings suggest that melatonin may act as a promising therapeutic agent for reducing post-stroke neuroinflammation by targeting HMGB1 and the subsequent signaling axis.
Collapse
Affiliation(s)
- Enkhmurun Chibaatar
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Kai Le
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Idriss Ali Abdoulaye
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Shanshan Wu
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.,School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, China
| | - Yijing Guo
- Department of Neurology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, Jiangsu Province, China.
| |
Collapse
|
18
|
Paudel YN, Angelopoulou E, Piperi C, Othman I, Shaikh MF. HMGB1-Mediated Neuroinflammatory Responses in Brain Injuries: Potential Mechanisms and Therapeutic Opportunities. Int J Mol Sci 2020; 21:ijms21134609. [PMID: 32610502 PMCID: PMC7370155 DOI: 10.3390/ijms21134609] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 12/14/2022] Open
Abstract
Brain injuries are devastating conditions, representing a global cause of mortality and morbidity, with no effective treatment to date. Increased evidence supports the role of neuroinflammation in driving several forms of brain injuries. High mobility group box 1 (HMGB1) protein is a pro-inflammatory-like cytokine with an initiator role in neuroinflammation that has been implicated in Traumatic brain injury (TBI) as well as in early brain injury (EBI) after subarachnoid hemorrhage (SAH). Herein, we discuss the implication of HMGB1-induced neuroinflammatory responses in these brain injuries, mediated through binding to the receptor for advanced glycation end products (RAGE), toll-like receptor4 (TLR4) and other inflammatory mediators. Moreover, we provide evidence on the biomarker potential of HMGB1 and the significance of its nucleocytoplasmic translocation during brain injuries along with the promising neuroprotective effects observed upon HMGB1 inhibition/neutralization in TBI and EBI induced by SAH. Overall, this review addresses the current advances on neuroinflammation driven by HMGB1 in brain injuries indicating a future treatment opportunity that may overcome current therapeutic gaps.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| | - Iekhsan Othman
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Strength, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 47500, Malaysia;
- Correspondence: (Y.N.P.); (C.P.); (M.F.S.); Tel.: +6-01-8396-0285 (Y.N.P.); +30-210-746-2610 (C.P.); +60-3-5514-6000 (ext. 44483) or +60-3-5514-4483 (M.F.S.); Fax: +30-210-746-2703 (C.P.); +601-4283-2410 (M.F.S.)
| |
Collapse
|
19
|
Wan J, Huang L, Ji X, Yao S, Hamed Abdelaziz M, Cai W, Wang H, Cheng J, Dineshkumar K, Aparna V, Su Z, Wang S, Xu H. HMGB1-induced ILC2s activate dendritic cells by producing IL-9 in asthmatic mouse model. Cell Immunol 2020; 352:104085. [PMID: 32201004 DOI: 10.1016/j.cellimm.2020.104085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 01/20/2023]
Abstract
Asthma is a disease of the respiratory system that is commonly considered a T-helper 2 (Th2) cell-associated inflammatory disease. Group 2 innate lymphoid cells (ILC2s) promote the inflammatory responses in asthma by secreting type 2 cytokines. Interleukin (IL)-9 also serves as a promoting factor in asthma and it is well known that ILC2s have an autocrine effect of IL-9 to sustain their survival and proliferation. However, the specific role of ILC2-derived IL-9 in asthma remains unclear. HMGB1 (High-Mobility Group Box-1) is a nuclear protein, and Previous studies have shown that HMGB1 can regulate the differentiation of T-helper cells and participate in the development of asthma. But whether HMGB1 can regulate the innate lymphocytes in the pathological process of asthma is unknown. In this study we have shown increased presence of HMGB1 protein in the lung of mice with asthma, which was associated with increased secretion of IL-9 by ILC2s. This led to the activation of dendritic cells (DCs) that can accelerate the differentiation of Th2 cells and worsen the severity of asthma. Taken together, our study provides a complementary understanding of the asthma development and highlights a novel inflammatory pathway in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Jie Wan
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Lan Huang
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoyun Ji
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Shun Yao
- Center for Pituitary Tumor Surgery, Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | | | - Wei Cai
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Huixuan Wang
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Jianjun Cheng
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | | | - Vasudevan Aparna
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China
| | - Zhaoliang Su
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China; The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China; Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang 212001, China
| | - Huaxi Xu
- Department of Immunology, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
20
|
Kim H, Kim HJ, Kang JH, Kang BT, Yang MP. Evaluation of serum C-reactive protein and high mobility group box 1 concentrations in 22 dogs with acute pancreatitis: a pilot study. Vet Q 2020; 39:122-130. [PMID: 31401946 PMCID: PMC6830977 DOI: 10.1080/01652176.2019.1655178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Background: High mobility group box 1 (HMGB1) is an important mediator of systemic inflammatory response syndrome (SIRS) in humans with severe acute pancreatitis (AP), but there is little information regarding its role in dogs. Aim: To compare the serum concentrations of C-reactive protein (CRP) and HMGB1 in healthy dogs and those with AP with or without SIRS. Methods: The study included 22 dogs with AP and 20 healthy dogs. CRP and HMGB1 were assessed by ELISA. Statistical analyses were conducted by non-parametric tests. Results: Median (interquartile range) serum CRP and HMGB1 concentrations were significantly (P < 0.05) higher in dogs with AP [60.56 (14.50-140.10) µg/mL and 0.35 (0.03-1.12) ng/mL, respectively] than in healthy dogs [2.23 (1.75-5.14) µg/mL and 0.02 (0.01-0.05) ng/mL, respectively]. After the recommended treatments for AP, serum CRP concentration in AP dogs significantly decreased, but that of HMGB1 in AP dogs significantly increased. There was also a significant difference in median serum HMGB1 concentration between AP dogs with and without SIRS. The use of serum HMGB1 concentration of 0.35 ng/mL to distinguish AP dogs with and without SIRS was associated with a sensitivity of 87.5% and a specificity of 71.5%. A positive correlation was identified between HMGB1 and clinical severity of AP. All AP dogs had a positive outcome during hospitalization [6.0 (1.5-6.0) days]. Conclusion: Results indicate that HMGB1 might be a useful biomarker for the progression of AP and may play a role in progression of AP into SIRS in dogs.
Collapse
Affiliation(s)
- Hakhyun Kim
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University , Cheongju , Chungbuk , Republic of Korea
| | - Hyung-Jin Kim
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University , Cheongju , Chungbuk , Republic of Korea
| | - Ji-Houn Kang
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University , Cheongju , Chungbuk , Republic of Korea
| | - Byeong-Teck Kang
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University , Cheongju , Chungbuk , Republic of Korea
| | - Mhan-Pyo Yang
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University , Cheongju , Chungbuk , Republic of Korea
| |
Collapse
|
21
|
Qian QQ, Zhang X, Wang YW, Xu JW, Dong HQ, Li NN, Qian YN, Gui B. Pro-inflammatory role of high-mobility group box-1 on brain mast cells via the RAGE/NF-κB pathway. J Neurochem 2019; 151:595-607. [PMID: 31520526 DOI: 10.1111/jnc.14869] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 09/01/2019] [Accepted: 09/09/2019] [Indexed: 12/12/2022]
Abstract
High-mobility group box-1 (HMGB-1) acts as a pro-inflammatory cytokine contributing to the occurrence of many central inflammatory and infectious disorders. Brain mast cells (MCs) are the first responders to peripheral inflammatory stimulation because of their rapid response to external stimuli coupled with their release of preformed and newly synthesized reactive chemicals. Little is known about the involvement of brain MCs in the pro-inflammatory effects of HMGB-1 on the central nervous system (CNS). Thus, we investigated the activation process of MCs by HMGB-1 and explored whether this process is involved in the pro-inflammatory effects of HMGB-1 on the CNS. In this study, we used P815 cells to study the activating role of HMGB-1 on MCs and to explore its potential mechanism in vitro. In an in vivo study, adult male Sprague-Dawley rats received i.c.v. injection of sterile saline or cromoglycate (stabilizer of MCs) 30 min prior to i.p. injection of HMGB-1. Increased levels of tumor necrosis factor and IL-1β were observed in the P815 cells, as well as in the rats' brains, after HMGB-1 treatment. Pretreatment with the receptor of advanced glycation endproducts (RAGE)-siRNA inhibited the HMGB-1-induced inflammatory process in the P815 cells. Activation of the RAGE/nuclear factor-κB (NF-κB) pathway was observed in both the P815 cells and rats' brains. In addition, HMGB-1 induced the accumulation of brain MCs in the hippocampal CA1 region, and the blood-brain barrier was disrupted. Pretreatment with cromoglycate, a stabilizer of MCs, mitigated these HMGB-1-induced pro-inflammatory processes in rats. These findings indicate that brain MCs are involved in the pro-inflammatory effect of HMGB-1 on the CNS, probably via activating the RAGE/NF-κB pathway.
Collapse
Affiliation(s)
- Qing-Qing Qian
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yi-Wei Wang
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Jia-Wen Xu
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Hong-Quan Dong
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Na-Na Li
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yan-Ning Qian
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Bo Gui
- Department of Anesthesiology, 1st Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Targeted Temperature Management at 33°C or 36°C Produces Equivalent Neuroprotective Effects in the Middle Cerebral Artery Occlusion Rat Model of Ischemic Stroke. Shock 2019; 50:714-719. [PMID: 29337840 DOI: 10.1097/shk.0000000000001106] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Targeted temperature management (TTM, 32°C to 36°C) is one of the most successful achievements in modern resuscitation medicine. It has become standard treatment for survivors of sudden cardiac arrest to minimize secondary brain damage. TTM at 36°C is just as effective as TTM at 33°C and is actually preferred because it reduces adverse TTM-associated effects. TTM also likely has direct neuroprotective effects in ischemic brains in danger of stroke. It remains unclear, however, whether higher temperature TTM is equally effective in protecting the brain from the effects of stroke. Here, we asked whether TTM at 36°C is as effective as TTM at 33°C in improving outcomes in a middle cerebral artery occlusion (MCAO) model of ischemic stroke. After dividing rats randomly into MCAO, MCAO+33°C TTM, MCAO+36°C TTM, and sham groups, we subjected all of them except for the sham group to MCAO for 3 h (for the behavioral tests) or 4 h (for all other biochemical analyses). We found TTM protocols at both 33°C and 36°C to produce comparable reductions of infarct volumes in the MCAO territory and equally attenuate the extracellular release of high mobility group box 1 in postischemic brains. Both the TTM conditions prevent the mRNA induction of a major pro-inflammatory cytokine, tissue necrosis factor-α, in the ischemic penumbra region. Finally, both the TTM protocols produce similar improvements in neurological outcomes in rats, as measured by a battery of behavior tests 21 h after the start of reperfusion. These data acquired in a rat MCAO model suggest TTM at 36°C has excellent therapeutic potential for improving clinical outcomes for patients with acute ischemic stroke.
Collapse
|
23
|
Piotrowski J, Jędrzejewski T, Pawlikowska M, Wrotek S, Kozak W. High mobility group box 1 protein released in the course of aseptic necrosis of tissues sensitizes rats to pyrogenic effects of lipopolysaccharide. J Therm Biol 2019; 84:36-44. [PMID: 31466775 DOI: 10.1016/j.jtherbio.2019.05.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/24/2019] [Accepted: 05/31/2019] [Indexed: 12/16/2022]
Abstract
It is still an open question as to whether or not aseptic injuries affect the generation of fever due to exogenous pyrogens including bacterial products. Therefore, in the present paper we have investigated the course of endotoxin fever in rats induced with lipopolysaccharide (LPS; given intraperitoneally in a dose of 50 μg/kg) 48 h after subcutaneous administration of turpentine oil (TRP; 0.1 mL per rat) that causes aseptic necrosis of tissues. We found that febrile response was significantly augmented in the animals pre-treated with turpentine compared to control rats (pre-treated with saline), and that observed excessive elevation of body temperature (Tb) was accompanied by enhanced release of fever mediators: interleukin-6 (IL-6) and prostaglandin E2 (PGE2) into plasma. Moreover, we found that sensitization to pyrogenic effects of lipopolysaccharide was associated with the increase in plasma level of high mobility group box 1 protein (HMGB1), one of the best-known damage-associated molecular patterns (DAMP), which was recently discovered as inflammatory mediator. Since the injection of anti-HMGB1 antibodies weakened observed hyperpyrexia in the animals pre-treated with turpentine, we conclude that HMGB1 is a plasma-derived factor released in the course of aseptic injury that enhances pyrogenic effects of LPS.
Collapse
Affiliation(s)
- Jakub Piotrowski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland.
| | - Tomasz Jędrzejewski
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Małgorzata Pawlikowska
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Sylwia Wrotek
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| | - Wieslaw Kozak
- Department of Immunology, Faculty of Biology and Environment Protection, Nicolaus Copernicus University, 1 Lwowska Street, 87-100, Torun, Poland
| |
Collapse
|
24
|
Tripathi A, Shrinet K, Kumar A. HMGB1 protein as a novel target for cancer. Toxicol Rep 2019; 6:253-261. [PMID: 30911468 PMCID: PMC6416660 DOI: 10.1016/j.toxrep.2019.03.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 02/23/2019] [Accepted: 03/01/2019] [Indexed: 12/11/2022] Open
Abstract
Highly conserved nuclear protein High Mobility Group Box1 (HMGB1) present in mammals has functionality as an immuno-modulator in the form of cytokine molecule, as a nuclear factor to regulate these molecules and DNA structural determination. It has proximal homologous DNA binding domains Box-A, Box-B and distal C-terminal domain. Reduced form exists in basic condition has chemotaxis activity, while form with disulphide bond reduced at 106th cysteine showed cytokine activity. The oxidized form is devoid of both activities. HMGB1 binds and bends dsDNA and also activates genes for secretion of inflammatory cytokines such as IL-1β, TNF-α, IL-6 and IL-18. It can interact with transcription factors Rel/NF-κB and p53 responsible for up-regulating oncogenes. Oxidative stressed injured tissues actively secrete HMGB1 outside cells to necrotize other nearby tissues passively in cytosol. Acetylation of HMGB1 weakens its binding with DNA, and promotes its migration to different tissues leading to secretion of inflammatory-cytokines. HMGB1 expression has been found very important in the genesis and promotion of different cancer by promoting metastasis. In current article, we emphasized on condition based structural variability of HMGB1, mechanism of release, physiological functions and its functionality as a biomarker for cancer to be targeted to curb cancer genesis and progression.
Collapse
Affiliation(s)
| | | | - Arvind Kumar
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| |
Collapse
|
25
|
Aucott H, Lundberg J, Salo H, Klevenvall L, Damberg P, Ottosson L, Andersson U, Holmin S, Erlandsson Harris H. Neuroinflammation in Response to Intracerebral Injections of Different HMGB1 Redox Isoforms. J Innate Immun 2018; 10:215-227. [PMID: 29478057 DOI: 10.1159/000487056] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 01/20/2018] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Neuroinflammation triggered by infection or trauma is the cause of central nervous system dysfunction. High-mobility group box 1 protein (HMGB1), released from stressed and dying brain cells, is a potent neuroinflammatory mediator. The proinflammatory functions of HMGB1 are tightly regulated by post-translational redox modifications, and we here investigated detailed neuroinflammatory responses induced by the individual redox isoforms. METHODS Male Dark Agouti rats received a stereotactic injection of saline, lipopolysaccharide, disulfide HMGB1, or fully reduced HMGB1, and were accessed for blood-brain barrier modifications using magnetic resonance imaging (MRI) and inflammatory responses by immunohistochemistry. RESULTS AND CONCLUSIONS Significant blood-brain barrier disruption appeared 24 h after injection of lipopolysaccharide, disulfide HMGB1, or fully reduced HMGB1 compared to controls, as assessed in post-gadolinium T1-weighted MRI images and confirmed by increased uptake of FITC-conjugated dextran. Immunohistochemistry revealed that both HMGB1 isoforms also induced a local production of IL-1β. Additionally, disulfide HMGB1 increased major histocompatibility complex class II expression and apoptosis. Together, the results demonstrate that extracellular, cerebral HMGB1 causes significant blood-brain barrier disruption in a redox-independent manner and activates several components of neuroinflammation. Blocking HMGB1 might potentially improve clinical outcome in conditions such as stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Hannah Aucott
- Department of Medicine Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Lundberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Henna Salo
- Department of Medicine Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lena Klevenvall
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Peter Damberg
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Lars Ottosson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Andersson
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Helena Erlandsson Harris
- Department of Medicine Solna, Rheumatology Unit, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Andersson U, Yang H, Harris H. Extracellular HMGB1 as a therapeutic target in inflammatory diseases. Expert Opin Ther Targets 2018; 22:263-277. [PMID: 29447008 DOI: 10.1080/14728222.2018.1439924] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION High-mobility group box 1 (HMGB1) is a ubiquitous nuclear protein that promotes inflammation when released extracellularly after cellular activation, stress, damage or death. HMGB1 operates as one of the most intriguing molecules in inflammatory disorders via recently elucidated signal and molecular transport mechanisms. Treatments based on antagonists specifically targeting extracellular HMGB1 have generated encouraging results in a wide number of experimental models of infectious and sterile inflammation. Clinical studies are still to come. Areas covered: We here summarize recent advances regarding pathways for extracellular HMGB1 release, receptor usage, and functional consequences of post-translational modifications. The review also addresses results of preclinical HMGB1-targeted therapy studies in multiple inflammatory conditions and outlines the current status of emerging clinical HMGB1-specific antagonists. Expert opinion: Blocking excessive amounts of extracellular HMGB1, particularly the disulfide isoform, offers an attractive clinical opportunity to ameliorate systemic inflammatory diseases. Therapeutic interventions to regulate intracellular HMGB1 biology must still await a deeper understanding of intracellular HMGB1 functions. Future work is needed to create more robust assays to evaluate functional bioactivity of HMGB1 antagonists. Forthcoming clinical studies would also greatly benefit from a development of antibody-based assays to quantify HMGB1 redox isoforms, presently assessed by mass spectrometry methods.
Collapse
Affiliation(s)
- Ulf Andersson
- a Department of Women's and Children's Health, Center for Molecular Medicine (CMM) L8:04, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| | - Huan Yang
- b Laboratory of Biomedical Science , The Feinstein Institute for Medical Research , Manhasset , NY , USA
| | - Helena Harris
- c Unit of Rheumatology, Department of Medicine, Center for Molecular Medicine (CMM) L, 8:04, Karolinska Institutet , Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
27
|
An JY, Pang HG, Huang TQ, Song JN, Li DD, Zhao YL, Ma XD. AG490 ameliorates early brain injury via inhibition of JAK2/STAT3-mediated regulation of HMGB1 in subarachnoid hemorrhage. Exp Ther Med 2017; 15:1330-1338. [PMID: 29434719 PMCID: PMC5774435 DOI: 10.3892/etm.2017.5539] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Accepted: 02/27/2017] [Indexed: 11/06/2022] Open
Abstract
High mobility group box 1 (HMGB1) is a classic damage-associated molecular pattern that has an important role in the pathological inflammatory response. In vitro studies have demonstrated that the Janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway is involved in the regulation of HMGB1 expression, mediating the inflammatory response. Therefore, the purpose of the present study was to evaluate JAK2/STAT3 pathway involvement in the subarachnoid hemorrhage (SAH)-dependent regulation of HMGB1, using an in vivo rat model. A SAH model was established by endovascular perforation. Western blotting, immunohistochemistry and immunofluorescence were used to analyze HMGB1 expression after SAH. In addition, the effects of AG490 after SAH on JAK2/STAT3 phosphorylation, HMGB1 expression and brain damage were evaluated. The results of the present study demonstrated that JAK2/STAT3 was significantly phosphorylated (P<0.05) and the total HMGB1 protein level was significantly increased (P<0.05) after SAH. In addition, the cytosolic HMGB1 level after SAH demonstrated an initial increase followed by a decrease to the control level, while the nuclear HMGB1 level after SAH demonstrated the opposite trend, with an initial decrease and subsequent increase. AG490 administration after SAH significantly inhibited JAK2/STAT3 phosphorylation (P<0.05), suppressed the expression and translocation of HMGB1, reduced cortical apoptosis, brain edema and neurological deficits. These results demonstrated the involvement of the JAK2/STAT3 pathway in HMGB1 regulation after SAH.
Collapse
Affiliation(s)
- Ji-Yang An
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hong-Gang Pang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ting-Qin Huang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin-Ning Song
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dan-Dong Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yong-Lin Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xu-Dong Ma
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
28
|
Zenerino C, Nuzzo AM, Giuffrida D, Biolcati M, Zicari A, Todros T, Rolfo A. The HMGB1/RAGE Pro-Inflammatory Axis in the Human Placenta: Modulating Effect of Low Molecular Weight Heparin. Molecules 2017; 22:molecules22111997. [PMID: 29149067 PMCID: PMC6150179 DOI: 10.3390/molecules22111997] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/10/2017] [Accepted: 11/14/2017] [Indexed: 01/11/2023] Open
Abstract
We evaluated whether physiological and pre-eclamptic (PE) placentae, characterized by exacerbated inflammation, presented alterations in pro-inflammatory High Mobility Group Box 1 (HMGB1) and its Receptor of Advanced Glycation End products (RAGE) expression. Moreover, we investigated, in physiological placental tissue, the ability of Low Molecular Weight Heparin (LMWH) to modify HMGB1 structural conformation thus inhibiting RAGE binding and HMGB1/RAGE axis inflammatory activity. HMGB1, RAGE, IL-6 and TNFα (HMGB1/RAGE targets) mRNA expression were assessed by Real Time PCR. HMGB1, RAGE protein levels were assessed by western blot assay. Physiological term placental explants were treated by 0.5 U LMWH for 24 or 48 h. HMGB1 and RAGE expression and association were evaluated in LMWH explants by RAGE immunoprecipitation followed by HMGB1 immunoblot. HMGB1 spatial localization was evaluated by immuofluorescent staining (IF). HMGB1 expression was increased in PE relative to physiological placentae while RAGE was unvaried. 24 h LMWH treatment significantly up-regulated HMGB1 expression but inhibited HMGB1/RAGE complex formation in physiological explants. RAGE expression decreased in treated relative to untreated explants at 48 h. IF showed HMGB1 localization in both cytoplasm and nucleus of mesenchymal and endothelial cells but not in the trophoblast. IL-6 and TNFα gene expression were significantly increased at 24 h relative to controls, while they were significantly down-regulated in 48 h vs. 24 h LMWH explants. Our data depicted a new molecular mechanism through which LMWH exerts its anti-inflammatory effect on PE placentae, underlying the importance of HMGB1/RAGE axis in PE inflammatory response.
Collapse
Affiliation(s)
- Cristian Zenerino
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Anna Maria Nuzzo
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Domenica Giuffrida
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Marilisa Biolcati
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Alessandra Zicari
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy.
| | - Tullia Todros
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| | - Alessandro Rolfo
- Department of Surgical Sciences, University of Turin, 10126 Turin, Italy.
| |
Collapse
|
29
|
Kim SY, Koh WJ, Park HY, Jeon K, Lee SY, Yim JJ, Shin SJ. Down-Regulation of Serum High-Mobility Group Box 1 Protein in Patients with Pulmonary Tuberculosis and Nontuberculous Mycobacterial Lung Disease. Tuberc Respir Dis (Seoul) 2017; 80:153-158. [PMID: 28416955 PMCID: PMC5392486 DOI: 10.4046/trd.2017.80.2.153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/24/2016] [Accepted: 12/16/2016] [Indexed: 11/24/2022] Open
Abstract
Background Recently, increased levels of high-mobility group box 1 protein (HMGB1) have been identified in various inflammatory conditions and infections. However, no studies have evaluated the HMGB1 level in nontuberculous mycobacterial (NTM) lung disease, and compared it to mycobacterial lung disease. Methods A total of 60 patients newly diagnosed with NTM lung disease, 44 culture-positive pulmonary tuberculosis (TB) patients, and 34 healthy controls, were included in this study. The serum HMGB1 concentrations were quantified using HMGB1 enzyme-linked immunosorbent assay kits. Results Serum HMGB1 level in patients with pulmonary TB or NTM lung disease, was significantly lower than that of the healthy controls. In addition, the serum HMGB1 level in TB patients was significantly lower than patients with NTM lung disease. However, the levels in NTM patient subgroups did not differ according to the causative species, disease progression, and disease phenotype. Conclusion Although low levels of serum HMGB1 has the potential to be a marker of mycobacterial lung disease, these levels were unable to differentiate disease progression and disease phenotype in NTM lung diseases.
Collapse
Affiliation(s)
- Su-Young Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Won-Jung Koh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hye Yun Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyeongman Jeon
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soo-Youn Lee
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jae-Joon Yim
- Department of Internal Medicine and Lung Institute of Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Hendrix P, Foreman PM, Harrigan MR, Fisher WS, Vyas NA, Lipsky RH, Lin M, Walters BC, Tubbs RS, Shoja MM, Pittet JF, Mathru M, Griessenauer CJ. Impact of High-Mobility Group Box 1 Polymorphism on Delayed Cerebral Ischemia After Aneurysmal Subarachnoid Hemorrhage. World Neurosurg 2017; 101:325-330. [PMID: 28189859 DOI: 10.1016/j.wneu.2017.01.121] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 01/29/2017] [Accepted: 01/30/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVE The high-mobility group box 1 (HMGB1) protein is a eukaryotic, ubiquitously expressed protein that serves as a biomarker for various diseases and is involved in the promotion of a proinflammatory response to cell injury. In aneurysmal subarachnoid hemorrhage (aSAH), increased HMGB1 levels have been linked to poor outcome and an increased risk for cerebral vasospasm. The role of HMGB1 polymorphisms in aSAH has not been previously investigated. METHODS Patients with aSAH and controls enrolled in the prospective, 2-center CARAS (Cerebral Aneurysm Renin Angiotensin System) study were evaluated. The 3814 C/G HMGB1 single nucleotide polymorphism (SNP) rs2249825 was detected using 5'exonuclease (Taqman) genotyping assays from blood samples from patients with aSAH and controls. Associations between aSAH and its clinical sequelae with the HMGB1 SNP were assessed. RESULTS Samples from 149 patients with aSAH and 50 controls were available for analysis. No increased risk for aSAH associated with the SNP was found compared with the control group. Delayed cerebral ischemia (DCI) was defined as a cerebral infarction at the time of discharge from the intensive care unit and identified in 21.2% of patients with aSAH. In multivariable logistic regression analysis, the G allele of rs2249825 was independently associated with DCI (odds ratio, 5.695; 95% confidence interval, 1.804-17.975; P = 0.003). CONCLUSIONS The minor allele G of rs2249825 was associated with an increased risk for DCI, or cerebral infarction, after aSAH. This finding may be attributable to an increased HMGB1 protein expression in these patients.
Collapse
Affiliation(s)
- Philipp Hendrix
- Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine, Homburg/Saar, Germany.
| | - Paul M Foreman
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mark R Harrigan
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Winfield S Fisher
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nilesh A Vyas
- Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA
| | - Robert H Lipsky
- Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA; Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - Minkuan Lin
- Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - Beverly C Walters
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Neurosciences, Inova Health System, Falls Church, Virginia, USA; Department of Molecular Neuroscience, George Mason University, Fairfax, Virginia, USA
| | - R Shane Tubbs
- Seattle Science Foundation, Seattle, Washington, USA
| | - Mohammadali M Shoja
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jean-Francois Pittet
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Mali Mathru
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Christoph J Griessenauer
- Neurosurgical Service, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA; Department of Neurosurgery, Geisinger Health System, Danville, Pennsylvania, USA
| |
Collapse
|
31
|
A. Richard S, Min W, Su Z, Xu HX. Epochal neuroinflammatory role of high mobility group box 1 in central nervous system diseases. AIMS MOLECULAR SCIENCE 2017. [DOI: 10.3934/molsci.2017.2.185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
32
|
Weber MD, Godbout JP, Sheridan JF. Repeated Social Defeat, Neuroinflammation, and Behavior: Monocytes Carry the Signal. Neuropsychopharmacology 2017; 42:46-61. [PMID: 27319971 PMCID: PMC5143478 DOI: 10.1038/npp.2016.102] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/28/2016] [Accepted: 05/27/2016] [Indexed: 02/06/2023]
Abstract
Mounting evidence indicates that proinflammatory signaling in the brain affects mood, cognition, and behavior and is linked with the etiology of psychiatric disorders, including anxiety and depression. The purpose of this review is to focus on stress-induced bidirectional communication pathways between the central nervous system (CNS) and peripheral immune system that converge to promote a heightened neuroinflammatory environment. These communication pathways involve sympathetic outflow from the brain to the peripheral immune system that biases hematopoietic stem cells to differentiate into a glucocorticoid-resistant and primed myeloid lineage immune cell. In conjunction, microglia-dependent neuroinflammatory events promote myeloid cell trafficking to the brain that reinforces stress-related behavior, and is argued to play a role in stress-related psychiatric disorders. We will discuss evidence implicating a key role for endothelial cells that comprise the blood-brain barrier in propagating peripheral-to-central immune communication. We will also discuss novel neuron-to-glia communication pathways involving endogenous danger signals that have recently been argued to facilitate neuroinflammation under various conditions, including stress. These findings help elucidate the complex communication that occurs in response to stress and highlight novel therapeutic targets against the development of stress-related psychiatric disorders.
Collapse
Affiliation(s)
- Michael D Weber
- Division of Biosciences, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Division of Biosciences, The Ohio State University, 223 IBMR Building, 305 W 12th Avenue, 460 Medical Center Drive, Columbus, OH 43210, USA, Tel: 614-293-3392, Fax: 614-292-6087, E-mail:
| | - Jonathan P Godbout
- Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA,Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - John F Sheridan
- Division of Biosciences, The Ohio State University, Columbus, OH, USA,Center for Brain and Spinal Cord Repair, The Ohio State University, Columbus, OH, USA,Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
33
|
Lian YJ, Gong H, Wu TY, Su WJ, Zhang Y, Yang YY, Peng W, Zhang T, Zhou JR, Jiang CL, Wang YX. Ds-HMGB1 and fr-HMGB induce depressive behavior through neuroinflammation in contrast to nonoxid-HMGB1. Brain Behav Immun 2017; 59:322-332. [PMID: 27647532 DOI: 10.1016/j.bbi.2016.09.017] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/14/2016] [Accepted: 09/15/2016] [Indexed: 02/08/2023] Open
Abstract
High mobility group box 1 (HMGB1) has been implicated as a key factor in several neuroinflammatory conditions. Our previous study suggested that the release of central HMGB1 acts as a late-phase mediator in lipopolysaccharide (LPS)-induced depression. Recent findings indicate that the redox state of HMGB1 is a critical determinant of its immunomodulatory properties. Here, we aimed to investigate the potential mechanisms that link the redox states of HMGB1 to depression in mice. Distinct redox forms of recombinant HMGB1 (rHMGB1) were used that included fully reduced HMGB (fr-HMGB1), which acted as a chemokine, and disulfide-HMGB1 (ds-HMGB1), which possessed cytokine activity. Fr-HMGB1 in vivo was partially oxidized into ds-HMGB1; thus, the mutant protein non-oxidizable chemokine-HMGB (nonoxid-HMGB1) was applied. Concurrent with depressive behavior induced by four-week stress exposure, the HMGB1 concentrations in the serum and cerebral cortex substantially increased. Therefore, a single dose of rHMGB1 (200ng/5μl/mice) or vehicle was administered to mice via intracerebroventricular (i.c.v.) injection. The receptor inhibitors of TLR4/RAGE/CXCR4 (TAK-242/FPS-ZM1/AMD3100) (3mg/kg) were intraperitoneally injected 30min prior to rHMGB1 treatment. Depressive-like behavior was measured 20h post i.c.v. injection. Administration of fr-HMGB1 prolonged the immobility duration in the tail suspension test (TST) and decreased sucrose preference. In addition to depressive behavior, the hippocampal TNF-α protein slightly increased. These depressive behaviors and upregulation of hippocampal TNF-α were alleviated or abrogated by pretreatment with the inhibitors AMD3100, FPS-ZM1, and TAK-242. Alternatively, nonoxid-HMGB1 failed to induce TNF-α protein or prolong the immobility duration. As expected, ds-HMGB1 administration substantially upregulated hippocampal TNF-α protein, increased the immobility time in the TST and decreased sucrose preference. Moreover, both glycyrrhizin and TAK-242 improved ds-HMGB1-induced depressive behavior. Furthermore, TAK-242 significantly blocked the upregulation of hippocampal TNF-α protein and protected hippocampal myelin basic protein from ds-HMGB1-induced reduction. These drugs had no effect on the total or central distance in the open field test. Collectively, this initial experiment demonstrates the role and receptor mechanisms of HMGB1 under different redox states on the induction of depressive-like behavior. Both ds-HMGB1 and fr-HMGB1 may induce depressive-like behavior in vivo mainly via neuroinflammatory response activation.
Collapse
Affiliation(s)
- Yong-Jie Lian
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Hong Gong
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Teng-Yun Wu
- Team of Aviation Physical Examination, Air Force General Hospital of PLA, Beijing 100142, PR China
| | - Wen-Jun Su
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yi Zhang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yuan-Yuan Yang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Wei Peng
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Ting Zhang
- Department of Navy Medicine, Second Military Medical University, Shanghai 200433, PR China
| | - Jiang-Rui Zhou
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Chun-Lei Jiang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China
| | - Yun-Xia Wang
- Lab of Stress Medicine, Department of Psychology and Mental Health, Second Military Medical University, Shanghai 200433, PR China.
| |
Collapse
|
34
|
Wu HH, Liu YF, Yang SF, Lin WL, Chen SC, Han CP, Wang HL, Lin LY, Wang PH. Association of single-nucleotide polymorphisms of high-mobility group box 1 with susceptibility and clinicopathological characteristics of uterine cervical neoplasia in Taiwanese women. Tumour Biol 2016; 37:10.1007/s13277-016-5408-0. [PMID: 27704361 DOI: 10.1007/s13277-016-5408-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022] Open
Abstract
To date, no study associated the genetic polymorphisms of high-mobility group box 1 protein (HMGB1) with the development of uterine cervical cancer. We therefore conducted this study to investigate the associations of HMGB1 single-nucleotide polymorphisms (SNPs) with cervical carcinogenesis and clinicopathological characteristics of cancer patients. Five hundred two women, including 112 with invasive cancer, 85 with precancerous lesions of the uterine cervix, and 305 normal controls, were consecutively enrolled into this study. Analysis of HMGB1 SNPs was done by real-time polymerase chain reaction and genotyping. Our results found that the risk of susceptibility to cervical invasive cancer was 1.85 (95 % CI 1.12-3.04; p = 0.016) in women with TC and 1.99 (95 % CI 1.24-3.23; p = 0.005) in women with TC/CC after adjusting for age, using TT as a comparison reference in HMGB1 SNP rs1412125. In rs2249825, the increased risk was also seen for the development of cervical invasive cancer in women with CG [adjusted odds ratio (AOR) 2.04, 95 % CI 1.22-3.40; p = 0.006] or CG/GG (AOR 2.02, 95 % CI 1.22-3.32; p = 0.006) using CC as a comparison reference. An additional integrated in silico analysis confirmed that rs2249825 creates a binding site for v-Myb, which may affect HMGB1 expression. In conclusion, Taiwanese women with TC or TC/CC in HMGB1 SNP rs1412125 as well as CG or CG/GG in rs2249825 were susceptible to the development of cervical invasive cancer.
Collapse
Affiliation(s)
- Hsin-Hung Wu
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan
- Infertility Center, Xiamen EMBO Hospital Xiamen, Fujian, China
- Reproductive Medicine Center, Kinmen Hospital, Kinmen, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wea-Lung Lin
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shiuan-Chih Chen
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Family and Community Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chih-Ping Han
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Pathology, Chung Shan Medical University Hospital, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hsiang-Ling Wang
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan
- Department of Beauty Science, National Taichung University of Science and Technology, Taichung, Taiwan
| | - Long-Yau Lin
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, 110, Section 1, Chien-Kuo North Road, Taichung, 40201, Taiwan.
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
35
|
Harada S, Matsuura W, Liu K, Nishibori M, Tokuyama S. RETRACTED: Possible involvement of the HMGB1/RAGE signaling mechanism in the induction of central post-stroke pain induced by acute global cerebral ischemia. Brain Res 2016; 1646:433-440. [DOI: 10.1016/j.brainres.2016.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/06/2016] [Accepted: 06/18/2016] [Indexed: 01/03/2023]
|
36
|
Reiss CS. Innate Immunity in Viral Encephalitis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7153449 DOI: 10.1007/978-3-319-33189-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|
37
|
PPARγ-Dependent and -Independent Inhibition of the HMGB1/TLR9 Pathway by Eicosapentaenoic Acid Attenuates Ischemic Brain Damage in Ovariectomized Rats. J Stroke Cerebrovasc Dis 2015; 24:1187-95. [DOI: 10.1016/j.jstrokecerebrovasdis.2015.01.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/05/2015] [Indexed: 11/18/2022] Open
|
38
|
Role of high-mobility group box-1 in myocardial ischemia/reperfusion injury and the effect of ethyl pyruvate. Exp Ther Med 2015; 9:1537-1541. [PMID: 25780465 PMCID: PMC4353799 DOI: 10.3892/etm.2015.2290] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 01/29/2015] [Indexed: 12/26/2022] Open
Abstract
High-mobility group box-1 (HMGB1) acts as a proinflammatory cytokine that triggers and amplifies the inflammation cascade following ischemia/reperfusion (I/R). Ethyl pyruvate (EP) has been reported to inhibit HMGB1 release in several I/R models. This study was designed to investigate the potential role of HMGB1 in a rat myocardial I/R model and to determine the effect of EP. Male Sprague Dawley rats were subjected to 30 min myocardial ischemia and 48 h reperfusion. In protocol 1, the rats were assigned to one of four groups (n=16 per group): Phosphate-buffered saline (PBS) or recombinant human HMGB1 (rhHMGB1) at three different doses (1, 10 or 100 μg/kg). In protocol 2, the rats were also assigned to one of four groups (n=16 per group): Sham, control, EP and EP + rhHMGB1. EP (40 mg/kg) or rhHMGB1 (100 μg/kg) was injected intravenously prior to reperfusion. Hemodynamic measurements were performed, and myocardial infarct size (IS) was calculated. Western blotting was conducted to evaluate HMGB1, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) expression levels. In the protocol 1 rats, the IS was markedly increased in the rhHMGB1 (100 μg/kg) group compared with that in the PBS group, and this increase was accompanied by elevated levels of TNF-α and IL-6. In the protocol 2 rats, I/R resulted a 4.8-fold increase in HMGB1 expression with an increased IS and impaired cardiac function compared with the sham group. EP significantly inhibited the elevated HMGB1 level, suppressed the activated TNF-α and IL-6 and reduced cardiac dysfunction. This cardioprotection was abolished by rhHMGB1. In conclusion, accumulation of HMGB1 is deleterious to the heart following myocardial I/R. EP can exert a strong protective effect against myocardial I/R injury, and these benefits are associated with a reduction in HMGB1.
Collapse
|
39
|
Agalave NM, Svensson CI. Extracellular high-mobility group box 1 protein (HMGB1) as a mediator of persistent pain. Mol Med 2015; 20:569-78. [PMID: 25222915 DOI: 10.2119/molmed.2014.00176] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/09/2014] [Indexed: 12/30/2022] Open
Abstract
Although originally described as a highly conserved nuclear protein, high-mobility group box 1 protein (HMGB1) has emerged as a danger-associated molecular pattern molecule protein (DAMP) and is a mediator of innate and specific immune responses. HMGB1 is passively or actively released in response to infection, injury and cellular stress, providing chemotactic and cytokine-like functions in the extracellular environment, where it interacts with receptors such as receptor for advanced glycation end products (RAGE) and several Toll-like receptors (TLRs). Although HMGB1 was first revealed as a key mediator of sepsis, it also contributes to a number of other conditions and disease processes. Chronic pain arises as a direct consequence of injury, inflammation or diseases affecting the somatosensory system and can be devastating for the affected patients. Emerging data indicate that HMGB1 is also involved in the pathology of persistent pain. Here, we give an overview of HMGB1 as a proinflammatory mediator, focusing particularly on the role of HMGB1 in the induction and maintenance of hypersensitivity in experimental models of pain and discuss the therapeutic potential of targeting HMGB1 in conditions of chronic pain.
Collapse
Affiliation(s)
- Nilesh M Agalave
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Role of Extracellular Damage-Associated Molecular Pattern Molecules (DAMPs) as Mediators of Persistent Pain. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:251-79. [DOI: 10.1016/bs.pmbts.2014.11.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
41
|
Allette YM, Due MR, Wilson SM, Feldman P, Ripsch MS, Khanna R, White FA. Identification of a functional interaction of HMGB1 with Receptor for Advanced Glycation End-products in a model of neuropathic pain. Brain Behav Immun 2014; 42:169-77. [PMID: 25014009 PMCID: PMC4560334 DOI: 10.1016/j.bbi.2014.06.199] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 06/25/2014] [Accepted: 06/27/2014] [Indexed: 10/25/2022] Open
Abstract
Recent studies indicate that the release of high mobility group box 1 (HMGB1) following nerve injury may play a central role in the pathogenesis of neuropathic pain. HMGB1 is known to influence cellular responses within the nervous system via two distinct receptor families; the Receptor for Advanced Glycation End-products (RAGE) and Toll-like receptors (TLRs). The degree to which HMGB1 activates a receptor is thought to be dependent upon the oxidative state of the ligand, resulting in the functional isoforms of all-thiol HMGB1 (at-HMGB1) acting through RAGE, and disufide HMGB1 (ds-HMGB1) interacting with TLR4. Though it is known that dorsal root ganglia (DRG) sensory neurons exposed to HMGB1 and TLR4 agonists can influence excitation, the degree to which at-HMGB1 signaling through neuronal RAGE contributes to neuropathic pain is unknown. Here we demonstrate that at-HMGB1 activation of nociceptive neurons is dependent on RAGE and not TLR4. To distinguish the possible role of RAGE on neuropathic pain, we characterized the changes in RAGE mRNA expression up to one month after tibial nerve injury (TNI). RAGE mRNA expression in lumbar dorsal root ganglion (DRG) is substantially increased by post-injury day (PID) 28 when compared with sham injured rodents. Protein expression at PID28 confirms this injury-induced event in the DRG. Moreover, a single exposure to monoclonal antibody to RAGE (RAGE Ab) failed to abrogate pain behavior at PID 7, 14 and 21. However, RAGE Ab administration produced reversal of mechanical hyperalgesia on PID28. Thus, at-HMGB1 activation through RAGE may be responsible for sensory neuron sensitization and mechanical hyperalgesia associated with chronic neuropathic pain states.
Collapse
Affiliation(s)
- Yohance M Allette
- Medical Science Training Program, Department of Anatomy, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Michael R Due
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sarah M Wilson
- Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Polina Feldman
- Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Matthew S Ripsch
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Rajesh Khanna
- Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, United States; Department of Biochemistry and Molecular Biology, United States
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, United States; Program in Medical Neurosciences, Paul and Carole Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
42
|
Zhang C, Li C, Jia S, Yao P, Yang Q, Zhang Y. High-mobility group box 1 inhibition alleviates lupus-like disease in BXSB mice. Scand J Immunol 2014; 79:333-7. [PMID: 24612327 DOI: 10.1111/sji.12165] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 02/26/2014] [Indexed: 01/01/2023]
Abstract
High-mobility group box 1 protein (HMGB1), a ubiquitous nuclear DNA-binding protein, functions as a potent proinflammatory factor. In this study, we evaluated the effects of HMGB1 inhibition on murine lupus using the lupus-prone model. We treated male BXSB mice with neutralizing anti-HMGB1 monoclonal antibody (HMGB1 mAb) from age 16 weeks to 26 weeks. The control group received the same amount of control IgG. Lupus-prone male BXSB mice treated with HMGB1mAb showed attenuated proteinuria, glomerulonephritis, circulating anti-dsDNA and immune complex deposition. Levels of serum IL-1β, IL-6, IL-17 and IL-18 were also significantly decreased by administration of HMGB1mAb in lupus-prone BXSB mice. HMGB1mAb treatment also decreased the caspase-1 activity in the kidneys of BXSB mice and reduced the mouse mortality. Our study supports that HMGB1 inhibition alleviates lupus-like disease in BXSB mice and might be a potential treatment option for human SLE.
Collapse
Affiliation(s)
- C Zhang
- Department of Rheumatology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | | | | | | | | | | |
Collapse
|
43
|
Walker FR, Nilsson M, Jones K. Acute and chronic stress-induced disturbances of microglial plasticity, phenotype and function. Curr Drug Targets 2014; 14:1262-76. [PMID: 24020974 PMCID: PMC3788324 DOI: 10.2174/13894501113149990208] [Citation(s) in RCA: 219] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 08/19/2013] [Accepted: 08/26/2013] [Indexed: 12/16/2022]
Abstract
Traditionally, microglia have been considered to act as macrophages of the central nervous system. While this concept still remains true it is also becoming increasingly apparent that microglia are involved in a host of non-immunological activities, such as monitoring synaptic function and maintaining synaptic integrity. It has also become apparent that microglia are exquisitely sensitive to perturbation by environmental challenges. The aim of the current review is to critically examine the now substantial literature that has developed around the ability of acute, sub-chronic and chronic stressors to alter microglial structure and function. The vast majority of studies have demonstrated that stress promotes significant structural remodelling of microglia, and can enhance the release of pro-inflammatory cytokines from microglia. Mechanistically, many of these effects appear to be driven by traditional stress-linked signalling molecules, namely corticosterone and norepinephrine. The specific effects of these signalling molecules are, however, complex as they can exert both inhibitory and suppressive effects on microglia depending upon the duration and intensity of exposure. Importantly, research has now shown that these stress-induced microglial alterations, rather than being epiphenomena, have broader behavioural implications, with the available evidence implicating microglia in directly regulating certain aspects of cognitive function and emotional regulation.
Collapse
Affiliation(s)
- Frederick Rohan Walker
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia.
| | | | | |
Collapse
|
44
|
Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage: from mechanisms to clinical translation. Prog Neurobiol 2013; 115:25-44. [PMID: 24291544 DOI: 10.1016/j.pneurobio.2013.11.003] [Citation(s) in RCA: 455] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 02/07/2023]
Abstract
Intracerebral hemorrhage (ICH) accounts for 10-15% of all strokes and is associated with high mortality and morbidity. Currently, no effective medical treatment is available to improve functional outcomes in patients with ICH. Potential therapies targeting secondary brain injury are arousing a great deal of interest in translational studies. Increasing evidence has shown that inflammation is the key contributor of ICH-induced secondary brain injury. Inflammation progresses in response to various stimuli produced after ICH. Hematoma components initiate inflammatory signaling via activation of microglia, subsequently releasing proinflammatory cytokines and chemokines to attract peripheral inflammatory infiltration. Hemoglobin (Hb), heme, and iron released after red blood cell lysis aggravate ICH-induced inflammatory injury. Danger associated molecular patterns such as high mobility group box 1 protein, released from damaged or dead cells, trigger inflammation in the late stage of ICH. Preclinical studies have identified inflammatory signaling pathways that are involved in microglial activation, leukocyte infiltration, toll-like receptor (TLR) activation, and danger associated molecular pattern regulation in ICH. Recent advances in understanding the pathogenesis of ICH-induced inflammatory injury have facilitated the identification of several novel therapeutic targets for the treatment of ICH. This review summarizes recent progress concerning the mechanisms underlying ICH-induced inflammation. We focus on the inflammatory signaling pathways involved in microglial activation and TLR signaling, and explore potential therapeutic interventions by targeting the removal of hematoma components and inhibition of TLR signaling.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yanchun Wang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, Johns Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - R Anne Stetler
- Center of Cerebrovascular Disease Research, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital & The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
45
|
Lee CC, Lai YT, Chang HT, Liao JW, Shyu WC, Li CY, Wang CN. Inhibition of high-mobility group box 1 in lung reduced airway inflammation and remodeling in a mouse model of chronic asthma. Biochem Pharmacol 2013; 86:940-9. [PMID: 23948063 DOI: 10.1016/j.bcp.2013.08.003] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 07/19/2013] [Accepted: 08/02/2013] [Indexed: 01/06/2023]
Abstract
The role of high-mobility group box 1 (HMGB1) in chronic allergic asthma is currently unclear. Both airway neutrophilia and eosinophilia and increase in HMGB1 expression in the lungs in our murine model of chronic asthma. Inhibition of HMGB1 expression in lung in ovalbumin (OVA)-immunized mice decreased induced airway inflammation, mucus formation, and collagen deposition in lung tissues. Analysis of the numbers of CD4(+) T helper (Th) cells in the mediastinal lymph nodes and lungs revealed that Th17 showed greater increases than Th2 cells and Th1 cells in OVA-immunized mice; further, the numbers of Th1, Th2, and Th17 cells decreased in anti-HMGB1 antibody (Ab)-treated mice. In OVA-immunized mice, TLR-2 and TLR-4 expression, but not RAGE expression, was activated in the lungs and attenuated after anti-HMGB1 Ab treatment. The results showed that increase in HMGB1 release and expression in the lungs could be an important pathological mechanism underlying chronic allergic asthma and HMGB1 might a potential therapeutic target for chronic allergic asthma.
Collapse
Affiliation(s)
- Chen-Chen Lee
- Department of Microbiology and Immunology, School of Medicine, China Medicine University, Taichung, Taiwan; Graduate Institute of Immunology, College of Medicine, China Medicine University, Taichung, Taiwan; Graduate Institute of Basic Medical Science, College of Medicine, China Medicine University, Taichung, Taiwan.
| | | | | | | | | | | | | |
Collapse
|
46
|
Arrigo T, Chirico V, Salpietro V, Munafò C, Ferraù V, Gitto E, Lacquaniti A, Salpietro C. High-mobility group protein B1: a new biomarker of metabolic syndrome in obese children. Eur J Endocrinol 2013; 168:631-8. [PMID: 23384711 DOI: 10.1530/eje-13-0037] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Obesity is associated with a chronic low-grade inflammation. High-mobility group box 1 protein (HMGB1) plays a key role in inflammation and immunostimulatory and chemotactic processes. The aim of the study was to assess the role of HMGB1 in obese children and to evaluate its diagnostic profile in identifying childhood obesity-related complications, such as the metabolic syndrome (MS). PATIENTS AND METHODS Sixty obese children were enrolled and compared with 40 healthy children (control). Homeostasis model assessment of insulin resistance (HOMA-IR), lipid profile, thyroid hormones, and pro- and anti-inflammatory peptides such as C-reactive protein (CRP), adiponectin, interleukin 6 (IL6), IL18, IL23, TNFα, resistin, and HMGB1 were evaluated. Receiver operating characteristics (ROC) analysis was employed to calculate the area under the curve (AUC) for HMGB1, IL6, and adiponectin to find the best cutoff values capable of identifying MS in obese children. RESULTS HMGB1 levels were statistically higher in obese patients than in the control group (19.4±6.8 vs 3.7±1.2 ng/ml; P<0.0001). In obese patients, IL18, IL6, and resistin levels were significantly high, while adiponectin levels were low. At multivariate analysis, HMGB1 was found to be independently correlated with BMI, IL23, IL6, free triiodothyronine, HDL, and HOMA-IR. At ROC analysis, HMGB1 showed higher sensitivity and specificity (AUC, 0. 992; sensitivity, 94.7%; specificity, 97.5%) than IL6 and adiponectin in identifying MS in obese children. CONCLUSION HMGB1 plays an important role in the inflammatory process associated with childhood obesity. This peptide may be an important diagnostic marker for obesity-related complications, such as MS.
Collapse
Affiliation(s)
- Teresa Arrigo
- Department of Pediatric Sciences, University of Messina, 98100 Messina, Italy
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Eosinophil-derived neurotoxin is elevated in patients with amyotrophic lateral sclerosis. Mediators Inflamm 2013; 2013:421389. [PMID: 23533305 PMCID: PMC3590756 DOI: 10.1155/2013/421389] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 12/24/2012] [Accepted: 01/10/2013] [Indexed: 12/11/2022] Open
Abstract
Background and Objectives. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by loss of motor neurons in the brainstem, motor cortex, and spinal cord. Oxidative stress and neuroinflammation have been implicated in the pathophysiology of ALS. Members of the family of damage-associated molecular patterns, including reactive oxygen species, high-mobility group box 1, and eosinophil-derived neurotoxin (EDN), may participate in pathological conditions. In this study, we aim to discover new biomarker for detecting ALS. Materials and Methods. We examined 44 patients with ALS, 41 patients with Alzheimer's disease, 41 patients with Parkinson's disease, and 44 healthy controls. The concentration of serum EDN was measured using an enzyme-linked immunosorbent assay. Results. EDN levels were significantly increased 2.17-fold in the serum of patients with ALS as compared with healthy controls (P < 0.05). No correlation between the levels of serum EDN and various clinical parameters of ALS was found. Moreover, the levels of serum EDN in patients with Parkinson's disease and Alzheimer's disease and healthy controls were similar. Conclusion. A higher level of serum EDN was found specifically in patients with ALS, indicating that EDN may participate in the pathophysiology of ALS.
Collapse
|
48
|
Pang Y, Xiao R, Liu X, Li Q. High-mobility-group family genes from Lampetra japonica reveal their early origin and molecular evolution in the vertebrate lineage. Genes Genomics 2012. [DOI: 10.1007/s13258-011-0234-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
49
|
Huang JM, Hu J, Chen N, Hu ML. Relationship between plasma high-mobility group box-1 levels and clinical outcomes of ischemic stroke. J Crit Care 2012; 28:792-7. [PMID: 23137435 DOI: 10.1016/j.jcrc.2012.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Revised: 09/09/2012] [Accepted: 10/01/2012] [Indexed: 11/25/2022]
Abstract
PURPOSE High-mobility group box-1 (HMGB1) is regarded as a central mediator of inflammation and involved in many inflammatory diseases. This study aimed to investigate impact of plasma HMGB1 level on 1-year clinical outcomes of ischemic stroke. METHODS Plasma HMGB1 levels of 338 patients were quantified by enzyme-linked immunosorbent assay. The end points were mortality and unfavorable outcome (modified Rankin Scale score>2) after 1 year. RESULTS Plasma HMGB1 level emerged as an independent predictor of 1-year clinical outcomes. Its prognostic value was similar to National Institutes of Health Stroke Scale score's. It improved prognostic value of National Institutes of Health Stroke Scale score. CONCLUSION Plasma HMGB1 level represents a novel biomarker for predicting 1-year clinical outcomes of ischemic stroke.
Collapse
Affiliation(s)
- Ju-Ming Huang
- Department of Neurology, The First Hospital of Jia Xing, Jiaxing, 314001, PR China.
| | | | | | | |
Collapse
|
50
|
Zhu XD, Chen JS, Zhou F, Liu QC, Chen G, Zhang JM. Relationship between plasma high mobility group box-1 protein levels and clinical outcomes of aneurysmal subarachnoid hemorrhage. J Neuroinflammation 2012; 9:194. [PMID: 22883976 PMCID: PMC3424135 DOI: 10.1186/1742-2094-9-194] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Accepted: 07/24/2012] [Indexed: 12/04/2022] Open
Abstract
Background High-mobility group box 1 (HMGB1), originally described as a nuclear protein that binds to and modifies DNA, is now regarded as a central mediator of inflammation by acting as a cytokine. However, the association of HMGB1 in the peripheral blood with disease outcome and cerebrovasospasm has not been examined in patients with aneurysmal subarachnoid hemorrhage. Methods In this study, 303 consecutive patients were included. Upon admission, plasma HMGB1 levels were measured by ELISA. The end points were mortality after 1 year, in-hospital mortality, cerebrovasospasm and poor functional outcome (Glasgow Outcome Scale score of 1 to 3) after 1 year. Results Upon admission, the plasma HMGB1 level in patients was statistically significantly higher than that in healthy controls. A multivariate analysis showed that the plasma HMGB1 level was an independent predictor of poor functional outcome and mortality after 1 year, in-hospital mortality and cerebrovasospasm. A receiver operating characteristic curve showed that plasma HMGB1 level on admission statistically significantly predicted poor functional outcome and mortality after 1 year, in-hospital mortality and cerebrovasospasm of patients. The area under the curve of the HMGB1 concentration was similar to those of World Federation of Neurological Surgeons (WFNS) score and modified Fisher score for the prediction of poor functional outcome and mortality after 1 year, and in-hospital mortality, but not for the prediction of cerebrovasospasm. In a combined logistic-regression model, HMGB1 improved the area under the curve of WFNS score and modified Fisher score for the prediction of poor functional outcome after 1 year, but not for the prediction of mortality after 1 year, in-hospital mortality, or cerebrovasospasm. Conclusions HMGB1 level is a useful, complementary tool to predict functional outcome and mortality after aneurysmal subarachnoid hemorrhage. However, HMGB1 determination does not add to the accuracy of prediction of the clinical outcomes.
Collapse
Affiliation(s)
- Xiang-Dong Zhu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310000, PR China.
| | | | | | | | | | | |
Collapse
|