1
|
Di Marco F, Cufaro MC, Damiani V, Dufrusine B, Pizzinato E, Di Ferdinando F, Sala G, Lattanzio R, Dainese E, Federici L, Ponsaerts P, De Laurenzi V, Cicalini I, Pieragostino D. Proteomic meta-analysis unveils new frontiers for biomarkers research in pancreatic carcinoma. Oncogenesis 2025; 14:3. [PMID: 39956821 PMCID: PMC11830788 DOI: 10.1038/s41389-025-00547-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/20/2024] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
Pancreatic carcinoma (PC) is the sixth leading cause of cancer death in both sexes in 2022, responsible for almost 5% of all cancer deaths worldwide; it is characterized by a poor prognosis since most patients present with an unresectable and metastatic tumor. To date, the decreasing trend in mortality rates related to the most common cancers has contributed to making pancreatic cancer a serious public health problem. In the last few years, scientific research has led to many advances in diagnostic approaches, perioperative management, radiotherapy techniques, and systemic therapies for advanced disease, but only with modest incremental progress in PC patient outcomes. Most of the causes of this high mortality are, unfortunately, late diagnosis and an important therapeutic resistance; for this reason, the most recent high-throughput proteomics technologies focus on the identification of novel biomarkers and molecular profiling to generate new insights in the study of PC, to improve diagnosis and prognosis and to monitor the therapies progress. In this work, we present and discuss the integration of results from different revised studies on protein biomarkers in a global proteomic meta-analysis to understand which path to pursue scientific research. In particular, cancer signaling, inflammatory response, and cell migration and signaling have emerged as the main pathways described in PC, as well as scavenging of free radicals and metabolic alteration concurrently highlighted new research insights on this disease. Interestingly, from the study of upstream regulators, some were found to be shared by collecting data relating to both biological fluid and tissue biomarkers, side by side: specifically, TNF, LPS, p38-MAPK, AGT, miR-323-5p, and miR-34a-5p. By integrating many biological components with their interactions and environmental relationships, it's possible to achieve an in-depth description of the pathological condition in PC and define correlations between concomitant symptoms and tumor genesis and progression. In conclusion, our work may represent a strategy to combine the results from different studies on various biological samples in a more comprehensive way.
Collapse
Affiliation(s)
- Federica Di Marco
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Maria Concetta Cufaro
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Verena Damiani
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Beatrice Dufrusine
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Erika Pizzinato
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Telematic University of "Leonardo Da Vinci", Torrevecchia Teatina, Chieti, Italy
| | - Fabio Di Ferdinando
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gianluca Sala
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Rossano Lattanzio
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Enrico Dainese
- Department of Bioscience and Technology for Food Agriculture and Environment, University of Teramo, Teramo, Italy
| | - Luca Federici
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerpen, Belgium
| | - Vincenzo De Laurenzi
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Damiana Pieragostino
- Centre for Advanced Studies and Technology (CAST), "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, "G. d' Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
2
|
Shakel Z, Costa Lima SA, Reis S. Strategies to make human skin models based on cellular senescence for ageing research. Ageing Res Rev 2024; 100:102430. [PMID: 39032611 DOI: 10.1016/j.arr.2024.102430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/25/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Human skin ageing is closely related to the ageing of the whole organism, and it's a continuous multisided process that is influenced not only by genetic and physiological factors but also by the cumulative impact of environmental factors. Currently, there is a scientific community need for developing skin models representing ageing processes to (i) enhance understanding on the mechanisms of ageing, (ii) discover new drugs for the treatment of age-related diseases, and (iii) develop effective dermo-cosmetics. Bioengineers worldwide are trying to reproduce skin ageing in the laboratory aiming to better comprehend and mitigate the senescence process. This review provides details on the main ageing molecular mechanisms and procedures to obtain in vitro aged skin models.
Collapse
Affiliation(s)
- Zinaida Shakel
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Porto, Portugal
| | - Sofia A Costa Lima
- LAQV, REQUIMTE, ICBAS, School of Medicine and Biomedical Sciences, University of Porto, Portugal.
| | - Salette Reis
- LAQV, REQUIMTE, Faculty of Pharmacy, University of Porto, Portugal
| |
Collapse
|
3
|
Xu L, An T, Jia B, Wu Q, Shen J, Jin J, Liu J, Li C. Histone deacetylase 3-specific inhibitor RGFP966 attenuates oxidative stress and inflammation after traumatic brain injury by activating the Nrf2 pathway. BURNS & TRAUMA 2024; 12:tkad062. [PMID: 38708192 PMCID: PMC11069425 DOI: 10.1093/burnst/tkad062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 05/07/2024]
Abstract
Background Oxidative stress (OS) and inflammatory reactions play pivotal roles in secondary brain injury after traumatic brain injury (TBI). Histone deacetylase 3 (HDAC3) controls the acetylation of histones and non-histones, which has a significant impact on the central nervous system's reaction to damage. This research determined the implications of RGFP966, a new and specific inhibitor of HDAC3, for the antioxidant (AO) systems mediated by nuclear factor erythroid2-related factor 2 (Nrf2) and the Nod-like receptor protein 3 (NLRP3) inflammasome in TBI. The study also studied the underlying mechanisms of RGFP966's actions. Our objective was to examine the impacts and underlying RGFP966 mechanisms in TBI. Methods In vitro, a rat cortical neuron OS model was induced by H2O2, followed by the addition of RGFP966 to the culture medium. Neurons were collected after 24 h for western blot (WB), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and 2'-7'-dichlorodihydrofluorescein diacetate staining. In vivo, RGFP966 (10 mg/kg) was administered post-TBI. Brain tissue water content and modified neurological severity scores were assessed 72 h post-injury. Cortical tissues surrounding the focal injury were subjected to western blot, TUNEL staining, Nissl staining and immunofluorescence/immunohistochemistry staining, and malondialdehyde level, hindered glutathione content and superoxide dismutase activity were measured. Serum was collected for the enzyme-linked immunosorbent assay. Nrf2-specific shRNA lentivirus was injected into the lateral ventricle of rats for 7 days, and cerebral cortex tissue was analyzed by WB and real-time polymerase chain reaction. Results During in vitro and in vivo experiments, RGFP966 suppressed HDAC3 expression, promoted Nrf2 nuclear translocation, activated downstream AO enzymes, mitigated excessive reactive oxygen species production and alleviated nerve cell apoptosis. RGFP966 effectively reduced brain edema and histological damage and enhanced neurological and cognitive function in rats with TBI. RGFP966 markedly inhibited NLRP3 inflammasome activation mediated by high-mobility group box 1 (HMGB1)/toll-like receptor 4 (TLR4). Nrf2 knockdown in TBI rats attenuated the AO and anti-inflammatory, neuroprotective impacts of RGFP966. Conclusions Overall, our findings demonstrate that RGFP966 can mitigate the first brain damage and neurological impairments in TBI. The underlying mechanism involves triggering the Nrf2-mediated AO system and negatively regulating the HMGB1/TLR4-mediated NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Lanjuan Xu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Tingting An
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Baohui Jia
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Qiong Wu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jinggui Shen
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jie Jin
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Jing Liu
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| | - Chengjian Li
- Department of Critical Care Medicine, Zhengzhou Central Hospital affiliated to Zhengzhou University, Zhengzhou, Henan Province 450001, China
| |
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
|
Teodorczyk-Injeyan JA, Khella H, Injeyan HS. Clinical Biomarker of Sterile Inflammation, HMGB1, in Patients with Chronic Non-Specific Low Back Pain: A Pilot Cross-Sectional Study. Life (Basel) 2023; 13:life13020468. [PMID: 36836824 PMCID: PMC9959829 DOI: 10.3390/life13020468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/24/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
The present study explores whether the inflammatory biomarker of sterile inflammation, high mobility box 1 (HMGB1), contributes to the inflammatory/nociceptive pathophysiology that characterizes chronic non-specific low back pain (LBP). Patients with chronic LBP (N = 10, >3 pain score on a 11-point Visual Analogue Scale, VAS) and asymptomatic participants (N = 12) provided peripheral blood (PB) samples. The proportion of classical CD14++ monocytes within PB leukocytes was determined by flow cytometry. The plasma and extracellular HMGB1 levels in unstimulated adherent cell (AC) cultures were measured using specific immunoassays. HMGB1 localization in ACs was assessed by immunofluorescent staining. The relative gene expression levels of tumor necrosis factor α (TNFα), interleukin-1 beta (IL-1β) and HMGB1 were determined by quantitative polymerase chain reaction (qRT-PCR) in relation to the pain intensity (11-point VAS scores) in patients with LBP. The extracellular release of HMGB1 in the LBP patient AC cultures was significantly elevated (p = 0.001) and accompanied by its relocation into the cytoplasm from the nuclei. The number of CD14++ monocytes in the patients' PB was significantly (p = 0.03) reduced, while the HMGB1 plasma levels remained comparable to those of the controls. The mRNA levels of TNFα, IL-1β and HMGB1 were overexpressed relative to the controls and those of HMGB1 and IL-1β were correlated with the VAS scores at a significant level (p = 0.01-0.03). The results suggest that HMGB1 may play an important role in the pathophysiology of chronic non-specific LBP.
Collapse
Affiliation(s)
- Julita A. Teodorczyk-Injeyan
- Graduate Education and Research Programs, Canadian Memorial Chiropractic College, Toronto, ON M2H 3J1, Canada
- Correspondence: ; Tel.: +1-647-805-2030
| | - Heba Khella
- Department of Clinical Education, Canadian Memorial Chiropractic College, Toronto, ON M2H 3J1, Canada
| | - H. Stephen Injeyan
- Graduate Education and Research Programs, Canadian Memorial Chiropractic College, Toronto, ON M2H 3J1, Canada
| |
Collapse
|
6
|
He Y, Zhang Y, Zhang J, Hu X. The Key Molecular Mechanisms of Sini Decoction Plus Ginseng Soup to Rescue Acute Liver Failure: Regulating PPARα to Reduce Hepatocyte Necroptosis? J Inflamm Res 2022; 15:4763-4784. [PMID: 36032938 PMCID: PMC9417306 DOI: 10.2147/jir.s373903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
Purpose This study aimed to investigate the improvement effect of Sini Decoction plus Ginseng Soup (SNRS) on the LPS/D-GalN-induced acute liver failure (ALF) mouse model and the molecular mechanism of the SNRS effect. Methods To study the protective effect of SNRS on ALF mice, the ICR mice were firstly divided into 4 groups: Control group (vehicle-treated), Model group (LPS/D-GalN), SNRS group (LPS/D-GalN+SNRS), and Silymarin group (LPS/D-GalN+Silymarin), the therapeutic drug was administered by gavage 48h, 24h before, and 10 min after LPS/D-GalN injection. On this basis, the peroxisome proliferator-activated receptor (PPAR) α agonist (WY14643) and inhibitor (GW6471) were added to verify whether the therapeutic mechanism of SNRS is related to its promoting effect on PPARα. The animals are grouped as follows: Control group (vehicle-treated), Model group (LPS/D-GalN+DMSO), SNRS group (LPS/D-GalN+SNRS+DMSO), Inhibitor group (LPS/D-GalN+GW6471), Agonist group (LPS/D-GalN+WY14643), and Inhibitor+SNRS group (LPS/D-GalN+GW6471+SNRS). Results The protective effect of SNRS on the ALF model is mainly reflected in the reduction of serum alanine aminotransaminase (ALT) and aspartate aminotransaminase (AST) as well as the ameliorated pathology of the liver tissue. The survival rate of ALF mice treated with SNRS was significantly increased. Further mechanism studies showed that SNRS significantly promoted the protein expression of PPARα and decreased the expression of necroptosis proteins (RIP3, MLKL, p-MLKL) in ALF mice. Reduced necroptosis resulted in decreased HMGB1 release, which in turn inhibited the activation of TLR4-JNK and NLRP3 inflammasome signaling pathways and the expression of NF-κB protein induced by LPS/D-GalN. The expression of CPT1A, a key enzyme involved in fatty acid β-oxidation, was found to be significantly up-regulated in the SNRS treated group, accompanied by an increased adenosine-triphosphate (ATP) level, which may be the relevant mechanism by which SNRS reduces necroptosis. Conclusion The potential therapeutic effect of SNRS on ALF may be through promoting the expression of PPARα and increasing the level of ATP in liver tissue, thereby inhibiting necroptosis of hepatocytes, reducing hepatocyte damage, and improving liver function.
Collapse
Affiliation(s)
- Ying He
- Department of Gastroenterology, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, People's Republic of China.,Department of College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Yang Zhang
- Department of Infectious Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Junli Zhang
- Department of College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Xiaoyu Hu
- Department of Infectious Disease, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| |
Collapse
|
7
|
Fischer S, Nasyrov E, Brosien M, Preissner KT, Marti HH, Kunze R. Self-extracellular RNA promotes pro-inflammatory response of astrocytes to exogenous and endogenous danger signals. J Neuroinflammation 2021; 18:252. [PMID: 34727934 PMCID: PMC8561902 DOI: 10.1186/s12974-021-02286-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/28/2021] [Indexed: 01/16/2023] Open
Abstract
Objective Astrocytes participate in the local innate immune response of the central nervous system. In response to stress such as ischemia, activated cells release endogenous factors known as damage-associated molecular patterns (DAMPs). Self-extracellular RNA (eRNA) is such a ubiquitous alarm signal. However, it is unclear whether eRNA is involved in the early acute phase of cerebral ischemia and is sufficient to sensitize astrocytes towards a DAMP or PAMP (pathogen-associated molecular pattern) reaction. Methods Pro-inflammatory activation upon eRNA stimulation was characterized in primary murine astrocyte cultures. In vivo, an experimental stroke model was used to localize and quantify eRNA in murine brain sections. Using primary cortical neurons and the mouse hippocampal neuronal cell line HT-22, neuronal RNA release upon stress conditions related to cerebral hypoxia/ischemia was analyzed. Results While low-dose eRNA alone did not promote pro-inflammatory activation of astrocytes in culture, it strongly enhanced the expression of pro-inflammatory cytokines in the presence of either Pam2CSK4, a synthetic PAMP molecule that mimics bacterial infection, or high mobility group box 1 (HMGB1), a prominent DAMP. Synergism of eRNA/Pam2CSK4 and eRNA/HMGB1 was prevented by blockage of the astroglial toll-like receptor (TLR)-2. Inhibition of NF-κB- and mitogen-activated protein kinase-dependent signaling pathways hampered eRNA/Pam2CSK4-mediated pro-inflammatory activation of astrocytes. In vivo, the amount of non-nuclear, presumably extracellular ribosomal RNA in close proximity to neurons significantly accumulated across the infarct core and peri-infarct areas that was accompanied by transcriptional up-regulation of various pro-inflammatory factors. Accordingly, the exposure of neurons to hypoxic/ischemic stress in vitro resulted in the release of eRNA, partly mediated by active cellular processes dependent on the cytosolic calcium level. Conclusion The DAMP signal eRNA can sensitize astrocytes as active players in cerebral innate immunity towards exogenous and endogenous activators of inflammation (PAMPs and DAMPs) in a synergistic manner via TLR2-NF-κB-dependent signaling mechanisms. These findings provide new insights into the pathogenesis of ischemic stroke and other inflammatory neurological disorders. Further studies will clarify whether administration of RNase in vivo may serve as an effective treatment for inflammatory brain pathologies. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02286-w.
Collapse
Affiliation(s)
- Silvia Fischer
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.
| | - Emil Nasyrov
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany.,Department of Ophthalmology, University Eye Hospital, University of Tübingen, Tübingen, Germany
| | - Monika Brosien
- German Center for Lung Research, Cardio-Pulmonary Institute, Universities of Giessen and Marburg Lung Center, Justus-Liebig-University, Giessen, Germany
| | - Klaus T Preissner
- Department of Biochemistry, Medical School, Justus-Liebig-University, Giessen, Germany.,Department of Cardiology, Medical School, Kerckhoff-Heart-Research-Institute, Justus-Liebig-University, Giessen, Germany
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, Heidelberg University, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Xu Z, Jin Y, Gao Z, Zeng Y, Du J, Yan H, Chen X, Ping L, Lin N, Yang B, He Q, Luo P. Autophagic degradation of CCN2 (cellular communication network factor 2) causes cardiotoxicity of sunitinib. Autophagy 2021; 18:1152-1173. [PMID: 34432562 DOI: 10.1080/15548627.2021.1965712] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Excessive macroautophagy/autophagy is one of the causes of cardiomyocyte death induced by cardiovascular diseases or cancer therapy, yet the underlying mechanism remains unknown. We and other groups previously reported that autophagy might contribute to cardiomyocyte death caused by sunitinib, a tumor angiogenesis inhibitor that is widely used in clinic, which may help to understand the mechanism of autophagy-induced cardiomyocyte death. Here, we found that sunitinib-induced autophagy leads to apoptosis of cardiomyocyte and cardiac dysfunction as the cardiomyocyte-specific Atg7-/+ heterozygous mice are resistant to sunitinib. Sunitinib-induced maladaptive autophagy selectively degrades the cardiomyocyte survival mediator CCN2 (cellular communication network factor 2) through the TOLLIP (toll interacting protein)-mediated endosome-related pathway and cardiomyocyte-specific knockdown of Ccn2 through adeno-associated virus serotype 9 (AAV9) mimics sunitinib-induced cardiac dysfunction in vivo, suggesting that the autophagic degradation of CCN2 is one of the causes of sunitinib-induced cardiotoxicity and death of cardiomyocytes. Remarkably, deletion of Hmgb1 (high mobility group box 1) inhibited sunitinib-induced cardiomyocyte autophagy and apoptosis, and the HMGB1-specific inhibitor glycyrrhizic acid (GA) significantly mitigated sunitinib-induced autophagy, cardiomyocyte death and cardiotoxicity. Our study reveals a novel target protein of autophagic degradation in the regulation of cardiomyocyte death and highlights the pharmacological inhibitor of HMGB1 as an attractive approach for improving the safety of sunitinib-based cancer therapy.
Collapse
Affiliation(s)
- Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Ying Jin
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Zizheng Gao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Yan Zeng
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Jiangxia Du
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Xueqin Chen
- Department of Oncology, Hangzhou First People's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Li Ping
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Nengming Lin
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R.China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China.,Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
9
|
Park MH, Choi JE, Kim JR, Bae YK. Immunohistochemical Expressions of Senescence-Associated Secretory Phenotype and Its Association With Immune Microenvironments and Clinicopathological Factors in Invasive Breast Cancer. Pathol Oncol Res 2021; 27:1609795. [PMID: 34267603 PMCID: PMC8276694 DOI: 10.3389/pore.2021.1609795] [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: 03/04/2021] [Accepted: 06/16/2021] [Indexed: 01/05/2023]
Abstract
This study was undertaken to investigate immunohistochemical expression of the senescence-associated secretory phenotype (SASP) in invasive breast cancer (IBC) tissues and to determine relationships between SASP positivity and tumor microenvironments and the clinicopathological characteristics of IBC. Immunohistochemistry for senescence markers, that is, high mobility group box-1 (HMGB1), p16, p15, and decoy receptor 2 (DCR2), was performed in tissue microarrays of 1140 IBC samples. Cases positive for at least one of these four markers were considered SASP-positive. Relations between SASP and tumor characteristics, including immune microenvironments (stromal tumor-infiltrating lymphocytes [sTILs] density and numbers of intraepithelial CD103-positive [iCD103 + ] lymphocytes) and clinical outcomes were retrospectively evaluated. HMGB1, p16, p15, or DCR2 was positive in 6.7%, 26.6%, 21.1%, and 26.5%, respectively, of the 1,140 cases. Six hundred and five (53.1%) cases were SASP positive, and SASP positivity was significantly associated with histologic grade 3, high-sTIL and iCD103 + lymphocyte counts, absence of ER or PR, and a high Ki-67 index. Although SASP did not predict breast cancer-specific survival (BCSS) or disease-free survival (DFS) in the entire cohort, SASP positivity in luminal A IBC was associated with poor BCSS and DFS. However, patients with SASP-positive TNBC showed better survival than those with SASP-negative TNBC. In multivariate analysis, SASP positivity was an independent prognostic factor in both luminal A IBC and TNBC, although the effect on prognosis was the opposite. In conclusion, SASP would be involved in the modulation of immune microenvironments and tumor progression in IBC, and its prognostic significance depends on molecular subtype.
Collapse
Affiliation(s)
- Min Hui Park
- Department of Pathology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Jung Eun Choi
- Department of Surgery, Division of Breast Surgery, Yeungnam University College of Medicine, Daegu, South Korea
| | - Jae-Ryong Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu, South Korea
| | - Young Kyung Bae
- Department of Pathology, Yeungnam University College of Medicine, Daegu, South Korea
| |
Collapse
|
10
|
Regulation of TREM1-Mediated Inflammation in Hepatocellular Carcinoma Cells. REPORTS 2021. [DOI: 10.3390/reports4020017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC), accounting for more than 90% of cases of primary liver cancer, is the third most common cause of cancer-related death worldwide. Chronic inflammation precedes the development of cirrhosis and HCC. TREM (triggering receptor expressed on myeloid cell)-1 is an inflammatory marker and amplifier of inflammation that signals through PI3K and ERK1/2 to activate transcription factors, resulting in increased secretion of pro-inflammatory cytokines, causing chronic inflammation and predisposing the liver to carcinogenesis. Thus, targeting TREM-1 in HCC might be a potential therapeutic target. A low level of vitamin D has been associated with chronic inflammation and poor prognosis in HCC. Thus, we evaluated the effect of vitamin D on TREM-1 expression in the HCC cell line. Additionally, the effects of high mobility group box-1, lipopolysaccharide, and transcription factor PU.1 on the expression of TREM-1 in normal liver cells and HCC cells have been investigated in the presence and absence of vitamin D. The results showed increased expression of TREM-1 in HCC cells and with IL-6, TNF-α, LPS, and rHMGB-1 and decreased expression with calcitriol. Calcitriol also attenuated the effect of IL-6, TNF-α, LPS, and rHMGB-1 on TREM-1. Calcitriol treatment attenuated the proliferation, migration, and invasion of HCC cells. These results (in vitro) provide molecular and biochemical evidence that calcitriol significantly attenuates the expression of mediators of inflammation, and thus might be used therapeutically together with conventional treatment to delay the progression of HCC. Additionally, the negative regulation of TREM-1 by PU.1 suggests PU.1 as a potential therapeutic target.
Collapse
|
11
|
Yilmaz N, Yelboga Z, Yilmaz Y, Demirpence O. High mobility group box-1 levels in schizophrenia: Potential biomarker of remission phase. J Med Biochem 2021; 40:295-301. [PMID: 34177374 PMCID: PMC8199597 DOI: 10.5937/jomb0-28108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/04/2020] [Indexed: 11/02/2022] Open
Abstract
Background: Schizophrenia is a chronic mental disorder, characterized byacute exacerbation and remission phases. Immune system has a role in the pathophysiology of schizophrenia. High mobility group box-1 (HMGB-1) is a macrophage secreted protein activating immune cells to produce cytokines. The aim of this study was to evaluate HMGB-1 levels among patients with schizophrenia both in acute exacerbation and remission phases. Methods: Consecutive schizophrenia patients in acute exacerbation and remission phases were enrolled and compared with each other and with age-sex matched healthy subjects. Patients were assessed with the Scale for the Assessment of Positive Symptoms (SAPS), Scale for the Assessment of Negative Symptoms (SANS), Brief Psychiatric Rating Scale (BPRS), Clinical Global Impression Scale (CGI). Results: Mean HMGB-1 levels were not significantly different in acute exacerbation phase versus remission phase schizophrenia patients (2.139±0.564 g/L vs. 2.326± 0.471 g/L, p=0.335) and both were individually higher than the control group (1.791±0.444 g/L, p=0.05 for acute exacerbation vs control, p=0.002 for remission vs control). In remission phase schizophrenic patients, HMGB-1 levels were positively correlated with Scale For The Assessment of Positive Symptoms (r=0.447, p=0.015) and BPRS (r=0.397, p=0.033) scores and HMGB-1 levels were independently associated with BPRS. Conclusions: Serum HMGB-1 levels were shown to be increased in patients with schizophrenia patients irrespective of phase, there were no differences between patients in acute exacerbation and remission phase in terms of biomarker and HMGB-1 levels were related to symptom severity according to psychiatric scales among patients in remission phase of schizophrenia.
Collapse
Affiliation(s)
- Nuryil Yilmaz
- Cumhuriyet University, Faculty of Medicine, Department of Psychiatry, Sivas, Turkey
| | - Zekeriya Yelboga
- Cumhuriyet University, Faculty of Medicine, Department of Psychiatry, Sivas, Turkey
| | - Yavuz Yilmaz
- Cumhuriyet University, Faculty of Medicine, Department of Psychiatry, Sivas, Turkey
| | - Ozlem Demirpence
- Cumhuriyet University, Faculty of Medicine, Department of Biochemistry, Sivas, Turkey
| |
Collapse
|
12
|
Radnaa E, Richardson LS, Sheller-Miller S, Baljinnyam T, de Castro Silva M, Kumar Kammala A, Urrabaz-Garza R, Kechichian T, Kim S, Han A, Menon R. Extracellular vesicle mediated feto-maternal HMGB1 signaling induces preterm birth. LAB ON A CHIP 2021; 21:1956-1973. [PMID: 34008619 PMCID: PMC8162392 DOI: 10.1039/d0lc01323d] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Preterm birth (PTB; <37 weeks of gestation) impacts ∼11% of all pregnancies and contributes to 1 million neonatal deaths worldwide annually. An understanding of the feto-maternal (F-M) signals that initiate birthing (parturition) at term is critical to design strategies to prevent their premature activation, resulting in PTB. Although endocrine and immune cell signaling are well-reported, fetal-derived paracrine signals capable of transitioning quiescent uterus to an active state of labor are poorly studied. Recent reports have suggested that senescence of the fetal amnion membrane coinciding with fetal growth and maturation generates inflammatory signals capable of triggering parturition. This is by increasing the inflammatory load at the feto-maternal interface (FMi) tissues (i.e., amniochorion-decidua). High mobility group box 1 protein (HMGB1), an alarmin, is one of the inflammatory signals released by senescent amnion cells via extracellular vesicles (exosomes; 40-160 nm). Increased levels of HMGB1 in the amniotic fluid, cord and maternal blood are associated with term and PTB. This study tested the hypothesis that senescent amnion cells release HMGB1, which is fetal signaling capable of increasing FMi inflammation, predisposing them to parturition. To test this hypothesis, exosomes from amnion epithelial cells (AECs) grown under normal conditions were engineered to contain HMGB1 by electroporation (eHMGB1). eHMGB1 was characterized (quantity, size, shape, markers and loading efficiency), and its propagation through FMi was tested using a four-chamber microfluidic organ-on-a-chip device (FMi-OOC) that contained four distinct cell types (amnion and chorion mesenchymal, chorion trophoblast and decidual cells) connected through microchannels. eHMGB1 propagated through the fetal cells and matrix to the maternal decidua and increased inflammation (receptor expression [RAGE and TLR4] and cytokines). Furthermore, intra-amniotic injection of eHMGB1 (containing 10 ng) into pregnant CD-1 mice on embryonic day 17 led to PTB. Injecting carboxyfluorescein succinimidyl ester (CFSE)-labeled eHMGB1, we determined in vivo kinetics and report that eHMGB1 trafficking resulting in PTB was associated with increased FMi inflammation. This study determined that fetal exosome mediated paracrine signaling can generate inflammation and induce parturition. Besides, in vivo functional validation of FMi-OOC experiments strengthens the reliability of such devices to test physiologic and pathologic systems.
Collapse
Affiliation(s)
- Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA. and Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Samantha Sheller-Miller
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Tuvshintugs Baljinnyam
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Mariana de Castro Silva
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Ananth Kumar Kammala
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Rheanna Urrabaz-Garza
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Talar Kechichian
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555-1062, USA.
| |
Collapse
|
13
|
Liu Y, Du J, Peng P, Cheng R, Lin J, Xu C, Yang H, Cui W, Mao H, Li Y, Geng D. Regulation of the inflammatory cycle by a controllable release hydrogel for eliminating postoperative inflammation after discectomy. Bioact Mater 2021; 6:146-157. [PMID: 32817921 PMCID: PMC7426539 DOI: 10.1016/j.bioactmat.2020.07.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/03/2020] [Accepted: 07/17/2020] [Indexed: 12/25/2022] Open
Abstract
Surgery is the final choice for most patients with intervertebral disc degeneration (IDD). Operation-caused trauma will cause inflammation in the intervertebral disc. Serious inflammation will cause tissue defects and induce tissue degeneration, IDD recurrence and the occurrence of other diseases. Therefore, we proposed a scheme to treat recurrence after discectomy by inhibiting inflammation with an aspirin (ASP)-loaded hydrogel to restore the mechanical stability of the spine and relieve local inflammation. ASP-liposomes (ASP-Lips) were incorporated into a photocrosslinkable gelatin-methacryloyl (GelMA) via mixing. This material can effectively alleviate inflammation by inhibiting the release of high mobility group box 1 (HMGB1) from the nucleus to the cytoplasm. We further assessed the expression of inflammatory cytokines, such as interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α), and degeneration-related factors, such as type II collagen (COL-2), Aggrecan, matrix metallopeptidases-3 (MMP-3), MMP-13, a disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS-4) and ADAMTS-5 in rat nucleus pulpous cells. The level of IDD was analyzed through H&E, safranin-O staining and immunohistochemistry in rabbit samples. In vitro, we found that ASP-Lip@GelMA treatment significantly decreased inflammatory cytokines, MMP-3 and -13, and ADAMTS-4 and -5 and up-regulated COL-2 and Aggrecan via the inhibited release of HMGB-1 from the nucleus. In vivo, ASP-Lip@GelMA can effectively inhibit inflammation of local tissue after disc surgery and fill local tissue defects. This composite hydrogel system is a promising way to treat the recurrence of IDD after surgery without persistent complications.
Collapse
Affiliation(s)
- Yu Liu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Jiacheng Du
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Peng Peng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Ruoyu Cheng
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, PR China
| | - Jiayi Lin
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Congxin Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Huilin Yang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Wenguo Cui
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, PR China
| | - Haiqing Mao
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| | - Yuling Li
- Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Second Road, Shanghai, 200025, PR China
- Department of Orthopaedics, Affiliated Hospital of North Sichuan Medical College, No.63 Wenhua Road, Nanchong, Sichuan, 637000, PR China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, PR China
| |
Collapse
|
14
|
Zanetti LC, Weinlich R. Necroptosis, the Other Main Caspase-Independent Cell Death. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1301:123-138. [PMID: 34370290 DOI: 10.1007/978-3-030-62026-4_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The past decades witnessed the discovery of novel modes of cell death, such as ferroptosis, pyroptosis and necroptosis, all of them presenting common necrotic traits. In this chapter, we revisit the early discoveries that unveiled necroptosis as a distinct cell death mechanism. We describe necroptosis, its main regulators and their role in maintaining cellular homeostasis and in the disease state. We conclude by discussing its phenotypic similarities with ferroptosis and the possible crosstalk between these pathways.
Collapse
Affiliation(s)
- Larissa C Zanetti
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil.
| | - Ricardo Weinlich
- Hospital Israelita Albert Einstein. Av. Albert Einstein, São Paulo, SP, Brazil
| |
Collapse
|
15
|
Leucine-rich alpha-2 glycoprotein 1, high mobility group box 1, matrix metalloproteinase 3 and annexin A1 as biomarkers of ulcerative colitis endoscopic and histological activity. Eur J Gastroenterol Hepatol 2020; 32:1106-1115. [PMID: 32483088 DOI: 10.1097/meg.0000000000001783] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE The LRG, HMGB1, MMP3 and ANXA1 proteins have been implicated in different inflammatory pathways in ulcerative colitis (UC), but their role as specific biomarkers of both endoscopic and histological activity has yet to be elucidated. In the present study, we aimed to evaluate the LRG1, HMGB1, MMP3 and ANXA1 as potential serum biomarkers for UC endoscopic and histological activity. METHODS This cross-sectional study included UC patients under 5-ASA, and healthy controls (HC) undergoing colonoscopy. Blood and biopsy samples were obtained and endoscopic Mayo sub-score (Ms) was recorded for the UC patients. Intramucosal calprotectin as a marker of histologic activity was evaluated in all biopsy samples and serum LRG1, HMGB1, MMP3 and ANXA1 levels were measured in the blood samples. RESULTS The HCs ANXA1 level was lower compared to that of the UC group [P = 0.00, area under the curve (AUC) = 0.881] and so was the HCs MMP3 level compared to that of patients (P = 0.00, AUC = 0.835). The HCs ANXA1 levels were also lower compared to these of the independent Ms groups, even to the Ms = 0 (P = 0.00, AUC = 0.913). UC endoscopic activity was associated with MMP3 levels (r = 0.54, P = 0.000) but not with ANXA1, LRG1 and HMGB1 levels CONCLUSION: Serum ANXA1 is a potential diagnostic biomarker of UC and serum MMP3 is a potential biomarker of UC endoscopic and histological activity.
Collapse
|
16
|
Czepiel J, Biesiada G, Pitera E, Wołkow PP, Michalak M, Garlicki A. Decreased Expression of the High Mobility Group Box 1 ( HMGB1) Gene in Peripheral Blood in Patients with Mild or Moderate Clostridioides difficile Infection. Microorganisms 2020; 8:microorganisms8081217. [PMID: 32796569 PMCID: PMC7464922 DOI: 10.3390/microorganisms8081217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/05/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Cytokines are mediators of inflammation induced in the course of Clostridioides difficile infection (CDI). High Mobility Group Box 1 (HMGB1) is a cytokine playing an important role in the pathogenesis of numerous inflammatory and autoimmune diseases. The aim of the study was to assess the HMGB1 gene expression in the course of CDI. We have performed a prospective case-control study- including 55 adult patients, among them 27 with CDI, who were hospitalized from October 2018 to February 2020 and 28 healthy volunteers. We assessed: a complete blood count with differential leukocyte count, blood creatinine, albumin, and C-reactive protein (CRP) levels. Then, the expression of the HMGB1 gene was evaluated using quantitative Real-Time PCR. Patients with CDI were found to have a significant increase in white blood cells (WBC), neutrophil count, and CRP levels, they also exhibited decreased levels of albumin compared with controls. The HMGB1 gene expression was significantly lower among patients with CDI compared with the control group and significantly, inversely correlated with CRP level in blood. In conclusion, we have observed a decreased expression of the HMGB1 gene in peripheral blood of patients with mild or moderate CDI, which hypothetically could reflect their diminished capability to fight the pathogen.
Collapse
Affiliation(s)
- Jacek Czepiel
- Department of Infectious and Tropical Diseases, Jagiellonian University Medical College, 30-688 Krakow, Poland; (G.B.); (A.G.)
- Correspondence: ; Tel./Fax: +48-124-002022/17
| | - Grażyna Biesiada
- Department of Infectious and Tropical Diseases, Jagiellonian University Medical College, 30-688 Krakow, Poland; (G.B.); (A.G.)
| | - Ewelina Pitera
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Krakow, Poland; (E.P.); (P.P.W.)
| | - Paweł P. Wołkow
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, 31-034 Krakow, Poland; (E.P.); (P.P.W.)
- Department of Pharmacology, Jagiellonian University Medical College, 31-531 Krakow, Poland
| | | | - Aleksander Garlicki
- Department of Infectious and Tropical Diseases, Jagiellonian University Medical College, 30-688 Krakow, Poland; (G.B.); (A.G.)
| |
Collapse
|
17
|
Wang H, Zhou XM, Wu LY, Liu GJ, Xu WD, Zhang XS, Gao YY, Tao T, Zhou Y, Lu Y, Wang J, Deng CL, Zhuang Z, Hang CH, Li W. Aucubin alleviates oxidative stress and inflammation via Nrf2-mediated signaling activity in experimental traumatic brain injury. J Neuroinflammation 2020; 17:188. [PMID: 32539839 PMCID: PMC7294631 DOI: 10.1186/s12974-020-01863-9] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 06/02/2020] [Indexed: 12/16/2022] Open
Abstract
Background Aucubin (Au), an iridoid glycoside from natural plants, has antioxidative and anti-inflammatory bioactivities; however, its effects on a traumatic brain injury (TBI) model remain unknown. We explored the potential role of Au in an H2O2-induced oxidant damage in primary cortical neurons and weight-drop induced-TBI in a mouse model. Methods In vitro experiments, the various concentrations of Au (50 μg/ml, 100 μg/ml, or 200 μg/ml) were added in culture medium at 0 h and 6 h after neurons stimulated by H2O2 (100 μM). After exposed for 12 h, neurons were collected for western blot (WB), immunofluorescence, and M29,79-dichlorodihydrofluorescein diacetate (DCFH-DA) staining. In vivo experiments, Au (20 mg/kg or 40 mg/kg) was administrated intraperitoneally at 30 min, 12 h, 24 h, and 48 h after modeling. Brain water content, neurological deficits, and cognitive functions were measured at specific time, respectively. Cortical tissue around focal trauma was collected for WB, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, Nissl staining, quantitative real time polymerase chain reaction (q-PCR), immunofluorescence/immunohistochemistry, and enzyme linked immunosorbent assay (ELISA) at 72 h after TBI. RNA interference experiments were performed to determine the effects of nuclear factor erythroid-2 related factor 2 (Nrf2) on TBI mice with Au (40 mg/kg) treatment. Mice were intracerebroventricularly administrated with lentivirus at 72 h before TBI establishment. The cortex was obtained at 72 h after TBI and used for WB and q-PCR. Results Au enhanced the translocation of Nrf2 into the nucleus, activated antioxidant enzymes, suppressed excessive generation of reactive oxygen species (ROS), and reduced cell apoptosis both in vitro and vivo experiments. In the mice model of TBI, Au markedly attenuated brain edema, histological damages, and improved neurological and cognitive deficits. Au significantly suppressed high mobility group box 1 (HMGB1)-mediated aseptic inflammation. Nrf2 knockdown in TBI mice blunted the antioxidant and anti-inflammatory neuroprotective effects of the Au. Conclusions Taken together, our data suggest that Au provides a neuroprotective effect in TBI mice model by inhibiting oxidative stress and inflammatory responses; the mechanisms involve triggering Nrf2-induced antioxidant system.
Collapse
Affiliation(s)
- Han Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China.,Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-Ming Zhou
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Ling-Yun Wu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Guang-Jie Liu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Wei-Dong Xu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xiang-Sheng Zhang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Yong-Yue Gao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Tao Tao
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Yan Zhou
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Yue Lu
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Juan Wang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Chu-Lei Deng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Zong Zhuang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China
| | - Chun-Hua Hang
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China.
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321# Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
18
|
Umahara T, Uchihara T, Hirao K, Shimizu S, Hashimoto T, Kohno M, Hanyu H. Essential autophagic protein Beclin 1 localizes to atherosclerotic lesions of human carotid and major intracranial arteries. J Neurol Sci 2020; 414:116836. [PMID: 32344218 DOI: 10.1016/j.jns.2020.116836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/23/2020] [Accepted: 04/12/2020] [Indexed: 01/23/2023]
Abstract
Macrophage autophagy has been shown to exert a protective role in atherosclerosis. Beclin 1 is an essential autophagic protein, and the Beclin-1-interacting complex promotes the formation of autophagosomes. However, the localization of Beclin 1 in human atherosclerotic lesions has not been clarified to date. We hence investigated the immunolocalization of Beclin 1 in atherosclerotic lesions of human carotid and major intracranial arteries. Furthermore, we investigated the colocalization of Beclin 1 with the 14-3-3 eta isoform and high mobility group box 1 (HMGB1). Beclin 1 was observed in the cytoplasm of many foamy macrophages located near to or in the periphery of lipid-rich necrotic cores. Beclin 1 colocalized with the 14-3-3 eta isoform in carotid plaques, and also colocalized with HMGB1 in carotid plaques. This is the first demonstration of Beclin 1 immunolocalization in human carotid and main cerebral artery plaques. We believe that our results will contribute towards understanding the role of autophagy in atherosclerosis and towards the prevention of stroke.
Collapse
Affiliation(s)
- Takahiko Umahara
- Department of Neurology, Mizuno Memorial Rehabilitation Hospital, 5-5-5 Nishiarai, Adachi-ku, Tokyo 123-0848, Japan; Departments of Geriatric Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan.
| | - Toshiki Uchihara
- Neurology Clinic with Neuromorphomics Laboratory, Nitobe-Memorial Nakano General Hospital, Tokyo 164-8607, Japan
| | - Kentaro Hirao
- Departments of Geriatric Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Soichiro Shimizu
- Departments of Geriatric Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Takao Hashimoto
- Departments of Neurosurgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Michihiro Kohno
- Departments of Neurosurgery, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Haruo Hanyu
- Departments of Geriatric Medicine, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan
| |
Collapse
|
19
|
Liu H, Liu W, Qiu H, Zou D, Cai H, Chen Q, Zheng C, Xu D. Salvianolic acid B protects against myocardial ischaemia-reperfusion injury in rats via inhibiting high mobility group box 1 protein expression through the PI3K/Akt signalling pathway. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:1527-1539. [PMID: 31853618 PMCID: PMC7351826 DOI: 10.1007/s00210-019-01755-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 10/23/2019] [Indexed: 01/20/2023]
Abstract
Salvianolic acid B (Sal B) has a significant protective effect on myocardial ischaemia-reperfusion (I/R) injury. Therefore, the aims of this study were to determine the effects of Sal B on myocardial ischaemic-reperfusion (I/R) injury in rats and to explore whether its underlying mechanism of cardioprotection occurs through activating the expression of the phosphoinositide 3-kinase/protein, kinase B (PI3K/Akt) and inhibiting the expression of high mobility group protein 1 (HMGB1). Ninety Sprague-Dawley rats were randomized into five groups: group 1 (sham-operated), group 2 (myocardial I/R), group 3 (low dose of Sal B+I/R), group 4 (high dose of Sal B+I/R), and group 5 (high dose of Sal B+I/R+LY294002, which is a specific PI3k inhibitor). All I/R rats received 30 min myocardial ischaemia followed by 24-h reperfusion. Cardiac function, infarct size, myocardial injury marker levels, inflammatory response and cardiomyocyte apoptosis as well as Bcl-2, Bax, P-Akt, HMGB1 and TLR4 expression were measured. In the current study, Sal B significantly ameliorated myocardial I/R injury in a dose-dependent manner, ameliorated cardiac function, reduced myocardial infarction size, decreased myocardial injury marker expression, decreased inflammatory responses, reduced apoptosis, activated PI3K/Akt expression and inhibited HMGB1 expression. However, all effects of Sal B were significantly reversed by LY294002. Overall, the present study indicated that Sal B attenuated myocardial I/R injury by activating PI3K/Akt and inhibiting the release of HMGB1 in rats.
Collapse
Affiliation(s)
- Hanqing Liu
- Cardiovascular Department, Guangzhou Hospital of integrated Traditional and West Medicine, Guangzhou, 510800, China
| | - Wei Liu
- Geriatrics Department, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, Zhejiang, China
| | - Huiliang Qiu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Dezhi Zou
- Emergency Department, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Huayang Cai
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Internal Medicine Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Qiuxiong Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Chaoyang Zheng
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Danping Xu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
- Cardiovascular Department, Guangdong Provincial Hospital of Chinese Medicine, 2nd Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
- Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, 510006, China.
| |
Collapse
|
20
|
Umahara T, Uchihara T, Hirao K, Shimizu S, Hashimoto T, Akimoto J, Kohno M, Hanyu H. Frontotemporal dementia-associated protein "phosphorylated TDP-43" localizes to atherosclerotic lesions of human carotid and main cerebral arteries. Histol Histopathol 2019; 35:159-167. [PMID: 31259382 DOI: 10.14670/hh-18-140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transactivation response DNA binding protein (TARDP) of 43 kDa (TDP-43) is a nuclear protein pivotal in RNA processing. Because phosphorylated (p) TDP-43 has been identified as a component of ubiquitin-positive and tau-negative inclusions in frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS), it is considered to play a major role in neurodegenerative processes. We investigated the immunolocalization of pTDP-43 in atherosclerotic lesions of human carotid and main cerebral arteries. Furthermore, we investigated the co-localization between pTDP-43 and 14-3-3 eta isoform or high mobility group box 1 (HMGB1). pTDP-43 localized in the cytoplasm of many foamy macrophages located in the periphery of lipid-rich necrotic cores, and in the cytoplasm of infiltrated smooth muscle cell-like cells. pTDP-43 co-localized the 14-3-3 eta isoform in carotid plaques. pTDP-43 also co-localized HMGB1. This is the first demonstration of pTDP-43 immunolocalization in human carotid and main cerebral artery plaques. We believe that demonstration of the localization of pTDP-43 in atherosclerotic lesions is important as this may contribute to the establishment of the clinical diagnostic imaging of FTLD and ALS using the pTDP-43 epitope. Moreover, this finding may be useful for further understanding the role of TDP in cell death.
Collapse
Affiliation(s)
- Takahiko Umahara
- Department of Neurology, Mizuno Memorial Rehabilitation Hospital, Nisharai, Adachi-ku, Tokyo, Japan. .,Department of Geriatric Medicine, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan.,Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Toshiki Uchihara
- Laboratory of Structural Neuropathology, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Neurology Clinic and Neuromorphomics Laboratory, Nitobe-Memorial Nakano General Hospital, Tokyo, Japan
| | - Kentaro Hirao
- Department of Geriatric Medicine, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Soichiro Shimizu
- Department of Geriatric Medicine, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Takao Hashimoto
- Department of Neurosurgery, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Jiro Akimoto
- Department of Neurosurgery, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Michihiro Kohno
- Department of Neurosurgery, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| | - Haruo Hanyu
- Department of Geriatric Medicine, Tokyo Medical University, Nishishinjuku, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
21
|
Gao F, Zhang S. Salicin inhibits AGE-induced degradation of type II collagen and aggrecan in human SW1353 chondrocytes: therapeutic potential in osteoarthritis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1043-1049. [PMID: 30942091 DOI: 10.1080/21691401.2019.1591427] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Feng Gao
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Shanyong Zhang
- Department of Spine Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
22
|
Weng J, Yu L, Chen Z, Su H, Yu S, Zhang Y, Lei X, Chen L, Cui Y, Huang Q, Jiang Y, Guo X. β-Catenin phosphorylation at Y654 and Y142 is crucial for high mobility group box-1 protein-induced pulmonary vascular hyperpermeability. J Mol Cell Cardiol 2019; 127:174-184. [DOI: 10.1016/j.yjmcc.2018.12.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 12/21/2018] [Accepted: 12/25/2018] [Indexed: 12/19/2022]
|
23
|
Liming S, Guixia L, Wenxin S, Guirong T. HMGB1 signaling blocking protects against carbapenem-resistant klebsiella pneumoniae in a murine model of infection. Exp Lung Res 2018; 44:263-271. [PMID: 30595050 DOI: 10.1080/01902148.2018.1505976] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF THE STUDY Pulmonary infection with Klebsiella pneumoniae (KP) and carbapenem-resistant Klebsiella pneumoniae (CRKP) significantly contribute to morbidity and mortality in pneumonia. Recent studies have indicated that High-Mobility Group Box 1 Protein (HMGB1) plays an important role in the prevention and treatment of pneumonia. However the role of HMGB1 in CRKP-induced pneumonia has not been addressed. Materials andMethods: In vivo, we successfully established the KP and CRKP-induced pneumonia mouse model. We then tested the anti-HMGB1 IgG prevents CRKP-induced pneumonia. RESULTS The mice treated with the anti-HMGB1 IgG ameliorated CRKP-induced pulmonary infiltration of inflammatory cells, dissemination of bacteria and the cytokine storm by suppressing the HMGB1 signaling pathways. CONCLUSION These results indicate that HMGB1 may be an important contributor in these changes of CRKP-induced pneumonia. Thus, HMGB1 may provide a therapeutic target for reducing bacterial infection and lung inflammation in CRKP pneumonia.
Collapse
Affiliation(s)
- Shi Liming
- a Department of Clinical Laboratory , Heze municipal Hospital , Heze , China
| | - Li Guixia
- a Department of Clinical Laboratory , Heze municipal Hospital , Heze , China
| | - Shi Wenxin
- b Department of Scientific Research and Teaching , Heze municipal Hospital , Heze , China
| | - Tian Guirong
- b Department of Scientific Research and Teaching , Heze municipal Hospital , Heze , China
| |
Collapse
|
24
|
Zhang H, Lu X, Liu Z, Du K. Rosuvastatin reduces the pro-inflammatory effects of adriamycin on the expression of HMGB1 and RAGE in rats. Int J Mol Med 2018; 42:3415-3423. [PMID: 30320373 PMCID: PMC6202077 DOI: 10.3892/ijmm.2018.3928] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 10/03/2018] [Indexed: 12/22/2022] Open
Abstract
Rosuvastatin has cardiac protective effects through its anti‑inflammatory effects. The nuclear protein high‑mobility group box 1 (HMGB1) can activate inflammatory pathways when released from dying cells. The present study aimed to investigate the effects of rosuvastatin in adriamycin (ADR)‑treated rats. Adult male rats were randomized to three groups: i) Control group, ii) ADR group, and iii) ADR+rosuvastatin group. Serum biochemical indices were measured using an enzyme‑linked immunosorbent assay. Cardiac function was assessed by echocardiography. The expression of HMGB1 and receptors for advanced glycation end products (RAGE) were assessed by reverse transcription‑quantitative polymerase chain reaction analysis, western blot analysis, and immunohistochemistry. Cytokines were measured using flow cytometry. Rosuvastatin improved the biochemical indices and cardiac morphology and alleviated the pathological lesions. In the ADR+rosuvastatin group, the mRNA and protein levels of HMGB1 and RAGE in the myocardium were significantly lower compared with those in the ADR group (both P<0.05). The results showed that rosuvastatin significantly reduced the levels of HMGB1 and RAGE in the myocardium of the ADR‑treated rats. These results suggest that the protective effects of rosuvastatin may be associated with attenuation of the HMGB1/RAGE‑mediated inflammatory response in ADR‑treated rats. Despite this protective effect of rosuvastatin in the present study, it did not improve cardiac function in terms of the diastolic left ventricular internal dimension, systolic left ventricular internal dimension, left ventricular ejection fraction and left ventricular fractional shortening; this may be due the observation duration being insufficient.
Collapse
Affiliation(s)
- Haiyan Zhang
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Xiang Lu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Zhengxia Liu
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Kang Du
- Department of Geriatrics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
25
|
Wang AS, Dreesen O. Biomarkers of Cellular Senescence and Skin Aging. Front Genet 2018; 9:247. [PMID: 30190724 PMCID: PMC6115505 DOI: 10.3389/fgene.2018.00247] [Citation(s) in RCA: 270] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/22/2018] [Indexed: 02/06/2023] Open
Abstract
Cellular senescence is an irreversible growth arrest that occurs as a result of different damaging stimuli, including DNA damage, telomere shortening and dysfunction or oncogenic stress. Senescent cells exert a pleotropic effect on development, tissue aging and regeneration, inflammation, wound healing and tumor suppression. Strategies to remove senescent cells from aging tissues or preneoplastic lesions can delay tissue dysfunction and lead to increased healthspan. However, a significant hurdle in the aging field has been the identification of a universal biomarker that facilitates the unequivocal detection and quantification of senescent cell types in vitro and in vivo. Mammalian skin is the largest organ of the human body and consists of different cell types and compartments. Skin provides a physical barrier against harmful microbes, toxins, and protects us from ultraviolet radiation. Increasing evidence suggests that senescent cells accumulate in chronologically aged and photoaged skin; and may contribute to age-related skin changes and pathologies. Here, we highlight current biomarkers to detect senescent cells and review their utility in the context of skin aging. In particular, we discuss the efficacy of biomarkers to detect senescence within different skin compartments and cell types, and how they may contribute to myriad manifestations of skin aging and age-related skin pathologies.
Collapse
Affiliation(s)
- Audrey S Wang
- Cell Ageing, Skin Research Institute of Singapore (SRIS), A∗STAR, Singapore, Singapore
| | - Oliver Dreesen
- Cell Ageing, Skin Research Institute of Singapore (SRIS), A∗STAR, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
26
|
Hong G, Zheng D, Zhang L, Ni R, Wang G, Fan GC, Lu Z, Peng T. Administration of nicotinamide riboside prevents oxidative stress and organ injury in sepsis. Free Radic Biol Med 2018; 123:125-137. [PMID: 29803807 PMCID: PMC6236680 DOI: 10.1016/j.freeradbiomed.2018.05.073] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 05/01/2018] [Accepted: 05/19/2018] [Indexed: 01/11/2023]
Abstract
AIMS Sepsis-caused multiple organ failure remains the major cause of morbidity and mortality in intensive care units. Nicotinamide riboside (NR) is a precursor of nicotinamide adenine dinucleotide (NAD+), which is important in regulating oxidative stress. This study investigated whether administration of NR prevented oxidative stress and organ injury in sepsis. METHODS Mouse sepsis models were induced by injection of lipopolysaccharides (LPS) or feces-injection-in-peritoneum. NR was given before sepsis onset. Cultured macrophages and endothelial cells were incubated with various agents. RESULTS Administration of NR elevated the NAD+ levels, and elicited a reduction of oxidative stress, inflammation and caspase-3 activity in lung and heart tissues, which correlated with attenuation of pulmonary microvascular permeability and myocardial dysfunction, leading to less mortality in sepsis models. These protective effects of NR were associated with decreased levels of plasma high mobility group box-1 (HMGB1) in septic mice. Consistently, pre-treatment of macrophages with NR increased NAD+ content and reduced HMGB1 release upon LPS stimulation. NR also prevented reactive oxygen species (ROS) production and apoptosis in endothelial cells induced by a conditioned-medium collected from LPS-treated macrophages. Furthermore, inhibition of SIRT1 by EX527 offset the negative effects of NR on HMGB1 release in macrophages, and ROS and apoptosis in endothelial cells. CONCLUSIONS Administration of NR prevents lung and heart injury, and improves the survival in sepsis, likely by inhibiting HMGB1 release and oxidative stress via the NAD+/SIRT1 signaling. Given NR has been used as a health supplement, it may be a useful agent to prevent organ injury in sepsis.
Collapse
Affiliation(s)
- Guangliang Hong
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China; Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5
| | - Dong Zheng
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5; Department of Medicine, Western University, London, Ontario, Canada N6A 4G5; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Lulu Zhang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Rui Ni
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5
| | - Grace Wang
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Guo-Chang Fan
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhongqiu Lu
- Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Tianqing Peng
- Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada N6A 4G5; Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada N6A 4G5; Department of Medicine, Western University, London, Ontario, Canada N6A 4G5; Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
27
|
Kim YM, Park EJ, Kim HJ, Chang KC. Sirt1 S-nitrosylation induces acetylation of HMGB1 in LPS-activated RAW264.7 cells and endotoxemic mice. Biochem Biophys Res Commun 2018; 501:73-79. [PMID: 29680657 DOI: 10.1016/j.bbrc.2018.04.155] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 11/29/2022]
Abstract
Excessive inflammation plays a detrimental role in endotoxemia. A recent study indicated that alarmins such as high mobility group box 1 (HMGB1) have drawn attention as therapeutic targets of sepsis. Post-translational modification (i.e., acetylation of lysine residues) of HMGB1 leads to the release of HMGB1 into the cellular space, operating as a warning signal that induces inflammation. Sirtuin 1 (SIRT1) has been shown to negatively regulate HMGB1 hyperacetylation and its extracellular release in sepsis. Therefore, we hypothesized that the S-nitrosylation (SNO) of SIRT1 may disrupt the ability of SIRT1 to negatively regulate the hyperacetylation of HMGB1. As long as the S-nitrosylation of SIRT1 occurs during septic conditions, it may worsen the situation. We found that the activity of SIRT1 decreased as the SNO-SIRT1 levels increased, resulting in HMGB1 release by LPS in RAW264.7 cells. Both the iNOS inhibitor (1400 W) and silencing iNOS significantly inhibited SNO-SIRT1, allowing increases in SIRT1 activity that decreased the HMGB1 release by LPS. SNAP, a NO donor, significantly increased both SNO-SIRT1 levels and the HMGB1 release that was accompanied by decreased sirt1 activity. However, sirtinol, a Sirt1 inhibitor, by itself decreased Sirt1 activity compared to that of the control, so that it did not affect already increased SNO-SIRT levels by SNAP. Most importantly, in lung tissues of LPS-endotoxic mice, significantly increased levels of SNO-SIRT were found, which was inhibited by 1400 W treatment. Plasma nitrite and HMGB1 levels were significantly higher than those in the sham controls, and the elevated levels were significantly lowered in the presence of 1400 W. We concluded that the S-nitrosylation of Sirt1 under endotoxic conditions may uninhibit the acetylation of HMGB1 and its extracellular release.
Collapse
Affiliation(s)
- Young Min Kim
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea
| | - Eun Jung Park
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea
| | - Hye Jung Kim
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea.
| | - Ki Churl Chang
- Department of Pharmacology, College of Medicine Gyeongsang National University and Institute of Health Sciences, Jinju 660-290, Republic of Korea.
| |
Collapse
|
28
|
Zhou H, Li Y, Gui H, Zhao H, Wu M, Li G, Li Y, Bai Z, Yin Z, Redmond HP, Wang J, Wang JH, Zhao Z. Antagonism of Integrin CD11b Affords Protection against Endotoxin Shock and Polymicrobial Sepsis via Attenuation of HMGB1 Nucleocytoplasmic Translocation and Extracellular Release. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:1771-1780. [PMID: 29343555 DOI: 10.4049/jimmunol.1701285] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/15/2017] [Indexed: 09/13/2023]
Abstract
High mobility group box 1 (HMGB1), a chromatin-binding nuclear protein, plays a critical role in sepsis by acting as a key "late-phase" inflammatory mediator. Integrin CD11b is essential for inflammatory cell activation and migration, thus mediating inflammatory responses. However, it is unclear whether CD11b participates in the development of sepsis. In this study, we report that CD11b contributes to LPS-induced endotoxin shock and microbial sepsis, as antagonism of CD11b with the CD11b blocking Ab or CD11b inhibitor Gu-4 protects mice against LPS- and microbial sepsis-related lethality, which is associated with significantly diminished serum HMGB1 levels. Consistent with this, CD11b-deficient mice were more resistant to microbial sepsis with a much lower serum HMGB1 level compared with wild-type mice. Pharmacological blockage and genetic knockdown/knockout of CD11b in murine macrophages hampered LPS-stimulated HMGB1 nucleocytoplasmic translocation and extracellular release. Furthermore, silencing CD11b interrupted the interaction of HMGB1 with either a nuclear export factor chromosome region maintenance 1 or classical protein kinase C and inhibited classical protein kinase C-induced HMGB1 phosphorylation, the potential underlying mechanism(s) responsible for CD11b blockage-induced suppression of HMGB1 nucleocytoplasmic translocation and subsequent extracellular release. Thus, our results highlight that CD11b contributes to the development of sepsis, predominantly by facilitating nucleocytoplasmic translocation and active release of HMGB1.
Collapse
Affiliation(s)
- Huiting Zhou
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| | - Yanhong Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Huan Gui
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - He Zhao
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Ming Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Gang Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Yiping Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Zhenjiang Bai
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China
| | - Zhimin Yin
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| | - H Paul Redmond
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork T12YN60, Ireland
| | - Jian Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou 215025, China;
| | - Jiang Huai Wang
- Department of Academic Surgery, University College Cork, Cork University Hospital, Cork T12YN60, Ireland
| | - Zhihui Zhao
- Jiangsu Province Key Laboratory for Molecular and Medicine Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210023, China; and
| |
Collapse
|
29
|
Davies JE, Apta BHR, Harper MT. Cross-reactivity of anti-HMGB1 antibodies for HMGB2. J Immunol Methods 2018; 456:72-76. [PMID: 29453955 PMCID: PMC5886380 DOI: 10.1016/j.jim.2018.02.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/26/2018] [Accepted: 02/13/2018] [Indexed: 12/17/2022]
Abstract
HMGB1 and HMGB2 are DNA-interacting proteins but can also have extracellular actions during inflammation. Despite their relatively high homology, they may have distinct roles, making it essential to be able to differentiate between the two. Here we examine the specificity of five commercially-available anti-HMGB1 antibodies. By Western blotting of recombinant proteins and HMGB1−/− mouse embryonic fibroblasts, we identified only one HMGB1 antibody that, under our experimental conditions, did not also detect HMGB2. Selecting specific antibodies for HMGB1 and HMGB2 allowed identification of distinct HMGB1 and HMGB2 subcellular pools in primary neutrophils.
Collapse
Affiliation(s)
- Jessica E Davies
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Bonita H R Apta
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
30
|
Ai P, Zhang X, Xie Z, Liu G, Liu X, Pan S, Wang H. The HMGB1 is increased in CSF of patients with an Anti-NMDAR encephalitis. Acta Neurol Scand 2018; 137:277-282. [PMID: 29023630 DOI: 10.1111/ane.12850] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is an autoimmune disorder of the central nervous system (CNS). Interleukin (IL)-6 and IL-17A may play important roles in the pathogenesis of this disease. High-mobility group box protein 1 (HMGB1), a small but highly conserved ubiquitous protein, is recognized to be a potent innate inflammatory mediator that can activate the nuclear factor light chain enhancer of activated B cells and release cytokines such as IL-6 and IL-17A when released extracellularly. However, whether cerebrospinal fluid (CSF) HMGB1 levels are altered in anti-NMDAR encephalitis is still unclear. OBJECTIVE The aim of this study was to determine whether a correlation exists between the CSF concentrations of HMGB1 and IL-6 and IL-17A in anti-NMDAR encephalitis patients. We also sought to assess whether HMGB1 influences the clinical outcomes in anti-NMDAR encephalitis patients. METHODS Thirty-three patients with anti-NMDAR antibodies and 38 controls were recruited. CSF HMGB1 was measured using an enzyme-linked immunosorbent assay. The main clinical outcomes were evaluated using the modified Rankin scale (mRS). The data were extracted using microarray analysis software. RESULTS AND CONCLUSION Our results showed significant increases in CSF HMGB1, IL-6, and IL-17A (P < .05) in anti-NMDAR encephalitis patients. But between 3 months' mRS scores in anti-NMDAR encephalitis patients and CSF data, there was no correlation. Our study suggests that HMGB1 CSF levels are increased in patients with anti-NMDAR encephalitis and reflect the underlying neuroinflammatory process.
Collapse
Affiliation(s)
- P. Ai
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - X. Zhang
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - Z. Xie
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - G. Liu
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - X. Liu
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - S. Pan
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| | - H. Wang
- Department of Neurology Nanfang Hospital Southern Medical University Guangzhou China
| |
Collapse
|
31
|
Di Ruscio M, Vernia F, Ciccone A, Frieri G, Latella G. Surrogate Fecal Biomarkers in Inflammatory Bowel Disease: Rivals or Complementary Tools of Fecal Calprotectin? Inflamm Bowel Dis 2017; 24:78-92. [PMID: 29272479 DOI: 10.1093/ibd/izx011] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Current noninvasive methods for assessing intestinal inflammation in inflammatory bowel disease (IBD) remain unsatisfactory. Along with C-reactive protein and erythrocyte sedimentation rate, fecal calprotectin (FC) is the standard test for assessing IBD activity, even though its specificity and accuracy are not optimal and it lacks a validated cutoff. Over the past few decades, several fecal markers released from intestinal inflammatory cells have been investigated in IBD; they are the subject of this systematic review. METHODS A systematic electronic search of the English literature up to April 2017 was performed using Medline and the Cochrane Library. Only papers written in English that analyzed fecal biomarkers in IBD were included. In vitro studies, animal studies, studies on blood/serum samples, and studies analyzing FC or fecal lactoferrin alone were excluded. RESULTS Out of 1023 citations, 125 eligible studies were identified. Data were grouped according to each fecal marker including S100A12, high-mobility group box 1, neopterin, polymorphonuclear neutrophil elastase, fecal hemoglobin, alpha1-antitrypsin, human neutrophil peptides, neutrophil gelatinase-associated lipocalin, chitinase 3-like-1, matrix metalloproteinase 9, lysozyme, M2-pyruvate kinase, myeloperoxidase, fecal eosinophil proteins, human beta-defensin-2, and beta-glucuronidase. Some of these markers showed a high sensitivity and specificity and correlated with disease activity, response to therapy, and mucosal healing. Furthermore, they showed a potential utility in the prediction of clinical relapse. CONCLUSIONS Several fecal biomarkers have the potential to become useful tools complementing FC in IBD diagnosis and monitoring. However, wide variability in their accuracy in assessment of intestinal inflammation suggests the need for further studies.
Collapse
Affiliation(s)
- Mirko Di Ruscio
- Gastroenterology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, L'Aquila, Italy
| | - Filippo Vernia
- Gastroenterology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, L'Aquila, Italy
| | - Antonio Ciccone
- Gastroenterology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, L'Aquila, Italy
| | - Giuseppe Frieri
- Gastroenterology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, L'Aquila, Italy
| | - Giovanni Latella
- Gastroenterology Unit, Department of Life, Health and Environmental Sciences, University of L'Aquila, Piazza S. Tommasi, Coppito, L'Aquila, Italy
| |
Collapse
|
32
|
Salidroside Inhibits HMGB1 Acetylation and Release through Upregulation of SirT1 during Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9821543. [PMID: 29333216 PMCID: PMC5733170 DOI: 10.1155/2017/9821543] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/25/2017] [Accepted: 07/25/2017] [Indexed: 12/24/2022]
Abstract
HMGB1, a highly conserved nonhistone DNA-binding protein, plays an important role in inflammatory diseases. Once released to the extracellular space, HMGB1 acts as a proinflammatory cytokine that triggers inflammatory reaction. Our previous study showed that salidroside exerts anti-inflammatory effect via inhibiting the JAK2-STAT3 signalling pathway. However, whether salidroside inhibits the release of HMGB1 is still unclear. In this study, we aim to study the effects of salidroside on HMGB1 release and then investigate the potential molecular mechanisms. In an experimental rat model of sepsis caused by CLP, salidroside administration significantly attenuated lung injury and reduced the serum HMGB1 level. In RAW264.7 cells, we investigated the effects of salidroside on LPS-induced HMGB1 release and then explored the underlying molecular mechanisms. We found that salidroside significantly inhibited LPS-induced HMGB1 release, and the inhibitory effect was correlated with the HMGB1 acetylation levels. Mechanismly, salidroside inhibits HMGB1 acetylation through the AMPK-SirT1 pathway. In addition, SirT1 overexpression attenuated LPS-induced HMGB1 acetylation and nucleocytoplasmic translocation. Furthermore, in SirT1 shRNA plasmid-transfected cells, salidroside treatment enhanced SirT1 expression and reduced LPS-activated HMGB1 acetylation and nucleocytoplasmic translocation. Collectively, these results demonstrated that salidroside might reduce HMGB1 release through the AMPK-SirT1 signalling pathway and suppress HMGB1 acetylation and nucleocytoplasmic translocation.
Collapse
|
33
|
Listyarifah D, Al-Samadi A, Salem A, Syaify A, Salo T, Tervahartiala T, Grenier D, Nordström DC, Sorsa T, Ainola M. Infection and apoptosis associated with inflammation in periodontitis: An immunohistologic study. Oral Dis 2017; 23:1144-1154. [DOI: 10.1111/odi.12711] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/16/2017] [Accepted: 06/28/2017] [Indexed: 02/01/2023]
Affiliation(s)
- D Listyarifah
- Department of Medicine, Clinicum; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
- Department of Dental Biomedical Sciences; Faculty of Dentistry; Universitas Gadjah Mada; Sleman Indonesia
| | - A Al-Samadi
- Department of Oral and Maxillofacial Diseases; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
| | - A Salem
- Department of Medicine, Clinicum; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
- Department of Oral and Maxillofacial Diseases; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
| | - A Syaify
- Department of Periodontology; Faculty of Dentistry; Universitas Gadjah Mada; Sleman Indonesia
| | - T Salo
- Department of Oral and Maxillofacial Diseases; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
- Department of Diagnostics and Oral Medicine; Institute of Dentistry; Oulu University Central Hospital; University of Oulu; Oulu Finland
| | - T Tervahartiala
- Department of Oral and Maxillofacial Diseases; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
| | - D Grenier
- Oral Ecology Research Group; Faculty of Dentistry; Université Laval; Quebec QC Canada
| | - DC Nordström
- Department of Internal Medicine and Rehabilitation; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
| | - T Sorsa
- Department of Oral and Maxillofacial Diseases; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
- Division of Periodontology; Department of Dental Medicine; Karolinska Institutet; Huddinge Sweden
| | - M Ainola
- Department of Medicine, Clinicum; University of Helsinki, and Helsinki University Central Hospital; Helsinki Finland
| |
Collapse
|
34
|
Shen W, Zhou J, Wang C, Xu G, Wu Y, Hu Z. High mobility group box 1 induces calcification of aortic valve interstitial cells via toll-like receptor 4. Mol Med Rep 2017; 15:2530-2536. [PMID: 28260034 PMCID: PMC5428883 DOI: 10.3892/mmr.2017.6287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 11/15/2016] [Indexed: 01/10/2023] Open
Abstract
Chronic inflammation and the calcification of aortic valve interstitial cells (AVICs) are the primary etiologies of calcific aortic valve disease (CAVD). However, the underlying mechanism remains to be elucidated. The present study investigated the importance of high mobility group box 1 (HMGB1) via toll-like receptor 4 (TLR4) for the regulation of inflammation and calcification in AVICs. It was determined that the expression levels of HMGB1 and TLR4 were increased in the calcific region of aortic valves with CAVD. In cultured primary AVICs from wild-type mice, HMGB1 treatment demonstrated a dose-dependent increase in mineralization levels and osteogenic gene expression. These effects were significantly reduced in AVICs obtained from TLR4 knockout mice (TLR4−/−). In addition, calcification was inhibited by TLR4-specific antibodies in primary AVICs. HMGB1 induced the activation of p38 and nuclear factor-κB (NF-κB) in TLR4−/− primary AVICs, and inhibited p38 and NF-κB in wild-type AVICs treated with TLR4-specific antibodies. The present study demonstrated that TLR4 may function as an essential mediator of HMGB1-induced calcification and in the activation of p38 and NF-κB.
Collapse
Affiliation(s)
- Wenjun Shen
- Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 310041, P.R. China
| | - Jianqing Zhou
- Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 310041, P.R. China
| | - Chaoyang Wang
- Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 310041, P.R. China
| | - Guangze Xu
- Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 310041, P.R. China
| | - Ying Wu
- Ningbo Medical Treatment Center, Lihuili Hospital, Ningbo, Zhejiang 310041, P.R. China
| | - Zhaohui Hu
- Department of Cardiovascular Disease, The Affiliated Tongji Hospital, Tongji University, Shanghai 210062, P.R. China
| |
Collapse
|
35
|
High Mobility Group Box-1 Protein and Outcomes in Critically Ill Surgical Patients Requiring Open Abdominal Management. Mediators Inflamm 2017; 2017:6305387. [PMID: 28286376 PMCID: PMC5329691 DOI: 10.1155/2017/6305387] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 12/19/2016] [Accepted: 01/22/2017] [Indexed: 12/20/2022] Open
Abstract
Background. Previous studies assessing various cytokines in the critically ill/injured have been uninformative in terms of translating to clinical care management. Animal abdominal sepsis work suggests that enhanced intraperitoneal (IP) clearance of Damage-Associated Molecular Patterns (DAMPs) improves outcome. Thus measuring the responses of DAMPs offers alternate potential insights and a representative DAMP, High Mobility Group Box-1 protein (HMGB-1), was considered. While IP biomediators are being recognized in critical illness/trauma, HMGB-1 behaviour has not been examined in open abdomen (OA) management. Methods. A modified protocol for HMGB-1 detection was used to examine plasma/IP fluid samples from 44 critically ill/injured OA patients enrolled in a randomized controlled trial comparing two negative pressure peritoneal therapies (NPPT): Active NPPT (ANPPT) and Barker's Vacuum Pack NPPT (BVP). Samples were collected and analyzed at the time of laparotomy and at 24 and 48 hours after. Results. There were no statistically significant differences in survivor versus nonsurvivor HMGB-1 plasma or IP concentrations at baseline, 24 hours, or 48 hours. However, plasma HMGB-1 levels tended to increase continuously in the BVP cohort. Conclusions. HMGB-1 appeared to behave differently between NPPT cohorts. Further studies are needed to elucidate the relationship of HMGB-1 and outcomes in septic/injured patients.
Collapse
|
36
|
Zhao J, Fuhrmann‐Stroissnigg H, Gurkar AU, Flores RR, Dorronsoro A, Stolz DB, St. Croix CM, Niedernhofer LJ, Robbins PD. Quantitative Analysis of Cellular Senescence in Culture and In Vivo. ACTA ACUST UNITED AC 2017; 79:9.51.1-9.51.25. [DOI: 10.1002/cpcy.16] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jing Zhao
- Department of Metabolism and Aging, The Scripps Research Institute Jupiter Florida
| | | | - Aditi U. Gurkar
- Department of Metabolism and Aging, The Scripps Research Institute Jupiter Florida
| | - Rafael R. Flores
- Department of Metabolism and Aging, The Scripps Research Institute Jupiter Florida
| | - Akaitz Dorronsoro
- Department of Metabolism and Aging, The Scripps Research Institute Jupiter Florida
| | - Donna B. Stolz
- Department of Cell Biology, University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
| | - Claudette M. St. Croix
- Department of Cell Biology, University of Pittsburgh School of Medicine Pittsburgh Pennsylvania
| | | | - Paul D. Robbins
- Department of Metabolism and Aging, The Scripps Research Institute Jupiter Florida
| |
Collapse
|
37
|
Richard SA, Min W, Su Z, Xu H. High Mobility Group Box 1 and Traumatic Brain Injury. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/jbbs.2017.72006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
38
|
Menon R, Behnia F, Polettini J, Saade GR, Campisi J, Velarde M. Placental membrane aging and HMGB1 signaling associated with human parturition. Aging (Albany NY) 2016; 8:216-30. [PMID: 26851389 PMCID: PMC4789578 DOI: 10.18632/aging.100891] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Aging is associated with the onset of several diseases in various organ systems; however, different tissues may age differently, rendering some of them dysfunctional sooner than others. Placental membranes (fetal amniochorionic membranes) protect the fetus throughout pregnancy, but their longevity is limited to the duration of pregnancy. The age-associated dysfunction of these membranes is postulated to trigger parturition. Here, we investigated whether cellular senescence-the loss of cell division potential as a consequence of stress-is involved in placental membrane function at term. We show telomere reduction, p38 MAPK activation, increase in p21 expression, loss of lamin B1 loss, increase in SA-β-galactosidase , and senescence-associated secretory phenotype (SASP) gene expression in placental membranes after labor and delivery (term labor [TL]) compared to membranes prior to labor at term (term, not-in-labor [TNIL]). Exposing TNIL placental membranes to cigarette smoke extract, an oxidative stress inducer, also induced markers of cellular senescence similar to those in TL placental membranes. Bioinformatics analysis of differentially expressed SASP genes revealed HMGB1 signaling among the top pathways involved in labor. Further, we show that recombinant HMGB1 upregulates the expression of genes associated with parturition in myometrial cells. These data suggest that the natural physiologic aging of placental tissues is associated with cellular senescence and human parturition.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Faranak Behnia
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Jossimara Polettini
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - George R Saade
- Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Department of Cell and Molecular Biology, Lawrence Berkley National Laboratory, Berkeley, CA 94720, USA
| | - Michael Velarde
- Buck Institute for Research on Aging, Novato, CA 94945, USA.,Institute of Biology, University of Philippines, Diliman, 1101 Quezon City, Philippines
| |
Collapse
|
39
|
Knapp DJ, Harper KM, Whitman BA, Zimomra Z, Breese GR. Stress and Withdrawal from Chronic Ethanol Induce Selective Changes in Neuroimmune mRNAs in Differing Brain Sites. Brain Sci 2016; 6:brainsci6030025. [PMID: 27472367 PMCID: PMC5039454 DOI: 10.3390/brainsci6030025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/10/2016] [Accepted: 07/20/2016] [Indexed: 12/12/2022] Open
Abstract
Stress is a strong risk factor in alcoholic relapse and may exert effects that mimic aspects of chronic alcohol exposure on neurobiological systems. With the neuroimmune system becoming a prominent focus in the study of the neurobiological consequences of stress, as well as chronic alcohol exposure proving to be a valuable focus in this regard, the present study sought to compare the effects of stress and chronic ethanol exposure on induction of components of the neuroimmune system. Rats were exposed to either 1 h exposure to a mild stressor (restraint) or exposure to withdrawal from 15 days of chronic alcohol exposure (i.e., withdrawal from chronic ethanol, WCE) and assessed for neuroimmune mRNAs in brain. Restraint stress alone elevated chemokine (C–C motif) ligand 2 (CCL2), interleukin-1-beta (IL-1β), tumor necrosis factor alpha (TNFα) and toll-like receptor 4 (TLR4) mRNAs in the cerebral cortex within 4 h with a return to a control level by 24 h. These increases were not accompanied by an increase in corresponding proteins. Withdrawal from WCE also elevated cytokines, but did so to varying degrees across different cytokines and brain regions. In the cortex, stress and WCE induced CCL2, TNFα, IL-1β, and TLR4 mRNAs. In the hypothalamus, only WCE induced cytokines (CCL2 and IL-1β) while in the hippocampus, WCE strongly induced CCL2 while stress and WCE induced IL-1β. In the amygdala, only WCE induced CCL2. Finally—based on the previously demonstrated role of corticotropin-releasing factor 1 (CRF1) receptor inhibition in blocking WCE-induced cytokine mRNAs—the CRF1 receptor antagonist CP154,526 was administered to a subgroup of stressed rats and found to be inactive against induction of CCL2, TNFα, or IL-1β mRNAs. These differential results suggest that stress and WCE manifest broad neuroimmune effects in brain depending on the cytokine and brain region, and that CRF inhibition may not be a relevant mechanism in non-alcohol exposed animals. Overall, these effects are complex in terms of their neuroimmune targets and neuroanatomical specificity. Further investigation of the differential distribution of cytokine induction across neuroanatomical regions, individual cell types (e.g., neuronal phenotypes and glia), severity of chronic alcohol exposure, as well as across differing stress types may prove useful in understanding differential mechanisms of induction and for targeting select systems for pharmacotherapeutic intervention in alcoholism.
Collapse
Affiliation(s)
- Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Kathryn M Harper
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Buddy A Whitman
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - Zachary Zimomra
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| | - George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Psychiatry, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- Department of Pharmacology, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
- UNC Neuroscience Center, The University of North Carolina at Chapel Hill, CB7178, Chapel Hill, NC 27599-7178, USA.
| |
Collapse
|
40
|
Wang Q, Liu GP, Xue FS, Wang SY, Cui XL, Li RP, Yang GZ, Sun C, Liao X. Combined Vagal Stimulation and Limb Remote Ischemic Perconditioning Enhances Cardioprotection via an Anti-inflammatory Pathway. Inflammation 2016; 38:1748-60. [PMID: 25772113 DOI: 10.1007/s10753-015-0152-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Various combined interventions to acquire enhanced cardioprotection are prevalent focuses of current research. This randomized experiment assessed whether combined vagal stimulation perconditioning (VSPerC) and limb remote ischemic perconditioning (LRIPerC) improved cardioprotection compared to the use of either treatment alone in an in vivo rat model of myocardial ischemia/reperfusion injury. A total of 100 male Sprague Dawley rats were randomly allocated into five groups: sham group, ischemia/reperfusion (IR) group, VSPerC group, LRIPerC group, and combined VSPerC and LRIPerC (COMPerC) group. Serum enzymatic markers, inflammatory cytokines, myocardial inflammatory cytokines, and infarct size were assessed. Infarct size decreased significantly in the COMPerC group compared to the VSPerC and LRIPerC groups. Serum intercellular adhesion molecule 1 (ICAM-1) level at 120 min of reperfusion, myocardial interleukin-1 (IL-1), ICAM-1, and tumor necrosis factor α (TNF-α) levels in the ischemic region decreased significantly in the COMPerC group compared to the VSPerC group, but myocardial IL-10 levels in the nonischemic region increased markedly in the COMPerC group. Serum TNF-α levels at 30, 60, and 120 min of reperfusion; serum IL-1, IL-6, ICAM-1, and high mobility group box-1 protein (HMGB-1) levels at 120 min of reperfusion; and myocardial IL-1, IL-6, ICAM-1, and TNF-α levels in the ischemic region decreased significantly in the COMPerC group compared to the LRIPerC group. However, myocardial IL-10 levels in both ischemic and nonischemic regions were evidently higher in the COMPerC group. This study concludes that combined VSPerC and LRIPerC enhances cardioprotection compared to either treatment alone. This result is likely attributable to a more potent regulation of inflammation.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Anesthesiology, Peking University People's Hospital, Beijing, China,
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Liu J, Zhang BL, Sun CL, Wang J, Li S, Wang JF. High mobility group box1 protein is involved in acute inflammation induced by Clostridium difficile toxin A. Acta Biochim Biophys Sin (Shanghai) 2016; 48:554-62. [PMID: 27151296 DOI: 10.1093/abbs/gmw038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/31/2016] [Indexed: 12/31/2022] Open
Abstract
High mobility group box1 (HMGB1), as a damage-associated inflammatory factor, contributes to the pathogenesis of numerous chronic inflammatory and autoimmune diseases. In this study, we explored the role of HMGB1 in CDI (Clostridium difficile infection) by in vivo and in vitro experiments. Our results showed that HMGB1 might play an important role in the acute inflammatory responses to C. difficile toxin A (TcdA), affect early inflammatory factors, and induce inflammation via the HMGB1-TLR4 pathway. Our study provides the essential information for better understanding the molecular mechanisms of CDI and the potential new therapeutic strategies for the treatment of this infection.
Collapse
Affiliation(s)
- Ji Liu
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Bei-Lei Zhang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Chun-Li Sun
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Jun Wang
- Shenzhen Huada Gene Research Institute, Shenzhen 518083, China
| | - Shan Li
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China Guangdong Province Key Laboratory of Fermentation and Enzyme Engineering, South China University of Technology, Guangzhou 510006, China
| | - Ju-Fang Wang
- School of Bioscience & Bioengineering, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
42
|
Breese GR, Knapp DJ. Persistent adaptation by chronic alcohol is facilitated by neuroimmune activation linked to stress and CRF. Alcohol 2016; 52:9-23. [PMID: 27139233 PMCID: PMC4855305 DOI: 10.1016/j.alcohol.2016.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/10/2015] [Accepted: 01/24/2016] [Indexed: 01/11/2023]
Abstract
This review updates the conceptual basis for the association of alcohol abuse with an insidious adaptation that facilitates negative affect during withdrawal from chronic intermittent alcohol (CIA) exposure - a change that later supports sensitization of stress-induced anxiety following alcohol abstinence. The finding that a CRF1-receptor antagonist (CRF1RA) minimized CIA withdrawal-induced negative affect supported an association of alcohol withdrawal with a stress mechanism. The finding that repeated stresses or multiple CRF injections into selected brain sites prior to a single 5-day chronic alcohol (CA) exposure induced anxiety during withdrawal provided critical support for a linkage of CIA withdrawal with stress. The determination that CRF1RA injection into positive CRF-sensitive brain sites prevented CIA withdrawal-induced anxiety provided support that neural path integration maintains the persistent CIA adaptation. Based upon reports that stress increases neuroimmune function, an effort was undertaken to test whether cytokines would support the adaptation induced by stress/CA exposure. Twenty-four hours after withdrawal from CIA, cytokine mRNAs were found to be increased in cortex as well as other sites in brain. Further, repeated cytokine injections into previously identified brain sites substituted for stress and CRF induction of anxiety during CA withdrawal. Discovery that a CRF1RA prevented the brain cytokine mRNA increase induced by CA withdrawal provided critical evidence for CRF involvement in this neuroimmune induction after CA withdrawal. However, the CRF1RA did not block the stress increase in cytokine mRNA increases in controls. The latter data supported the hypothesis that distinct mechanisms linked to stress and CA withdrawal can support common neuroimmune functions within a brain site. As evidence evolves concerning neural involvement in brain neuroimmune function, a better understanding of the progressive adaptation associated with CIA exposure will advance new knowledge that could possibly lead to strategies to combat alcohol abuse.
Collapse
Affiliation(s)
- George R Breese
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Pharmacology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Curriculum in Neurobiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; The UNC Neuroscience Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA.
| | - Darin J Knapp
- Bowles Center for Alcohol Studies, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA; Department of Psychiatry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7178, USA
| |
Collapse
|
43
|
Li S, Yin T, Li W, Yang J, Xu W, Zhou D. Association between follicular fluid levels of HMGB1 protein and outcomes in patients undergoing in vitro fertilization/intracytoplasmic sperm injection cycles. Exp Ther Med 2015; 9:1611-1616. [PMID: 26136867 DOI: 10.3892/etm.2015.2369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 02/10/2015] [Indexed: 12/31/2022] Open
Abstract
The aim of the present study was to evaluate the association between follicular fluid (FF) levels of high-mobility group box 1 (HMGB1) protein and the reproductive outcome in patients undergoing in vitro fertilization (IVF) with intracytoplasmic sperm injection (ICSI). FF samples were collected from the ovarian follicles (≥14 mm) of 143 infertile patients that had undergone IVF/ICSI, and the HMGB1 expression levels were determined using ELISA. Spearman's correlation and receiver operating characteristic (ROC) curve analysis were applied to analyze the results. Significantly increased levels of HMGB1 protein (7.38±2.02 vs. 6.14±2.52 ng/ml; P<0.01), endometrial thickness on the day of human chorionic gonadotropin (hCG) administration (10.3±1.3 vs. 9.7±1.7 mm; P<0.01) and retrieved oocyte counts (11.68±6.51 vs. 11.00±6.34; P<0.01) were observed in the pregnant group when compared with the non-pregnant group. Conversely, the level of luteinizing hormone on the day of hCG administration was significantly reduced in the pregnant group compared with the non-pregnant group (0.92±1.78 vs. 1.78±2.03 pmol/l, P<0.01). The ROC curve indicated a significant association between the FF level of HMGB1 protein and the pregnancy rate, with an area under the ROC curve of 0.673 (0.581-0.765; P<0.01). In addition, the HMGB1 protein level was shown to have a significant positive correlation with the endometrial thickness (r=0.170; P<0.05). Therefore, the present study indicated that the FF levels of HMGB1 protein are increased in pregnant patients and are positively correlated with endometrial thickness. Thus, FF levels of HMGB1 may be a useful factor for predicting the outcome of IVF/ICSI treatments.
Collapse
Affiliation(s)
- Saijiao Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tailang Yin
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Li
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wangming Xu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Danni Zhou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
44
|
Hu Z, Wang X, Gong L, Wu G, Peng X, Tang X. Role of high-mobility group box 1 protein in inflammatory bowel disease. Inflamm Res 2015; 64:557-63. [PMID: 26077468 DOI: 10.1007/s00011-015-0841-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 06/07/2015] [Accepted: 06/08/2015] [Indexed: 12/16/2022] Open
Abstract
High-mobility group box 1 (HMGB1) protein is a nuclear non-histone DNA-binding protein. It is released into the extracellular milieu and mediates inflammatory responses, which contribute to the pathogenesis of numerous inflammatory diseases, including inflammatory bowel disease (IBD). An online search was performed in PubMed and Web of Science databases for articles providing evidence on the role of HMGB1 in IBD. HMGB1 plays an important role in IBD pathogenesis. Application of HMGB1 antagonists reduced inflammatory reactions and ameliorated colitis in rodent models, which may provide new insights into the diagnosis and treatment of IBD.
Collapse
Affiliation(s)
- Zhen Hu
- Division of Gastroenterology, Wuxi No.2 Hospital Affiliated to Nanjing Medical University, Zhong Shan Road 68, Wuxi, Jiang Su, China
| | | | | | | | | | | |
Collapse
|
45
|
Liang Y, Hou C, Kong J, Wen H, Zheng X, Wu L, Huang H, Chen Y. HMGB1 binding to receptor for advanced glycation end products enhances inflammatory responses of human bronchial epithelial cells by activating p38 MAPK and ERK1/2. Mol Cell Biochem 2015; 405:63-71. [PMID: 25862459 DOI: 10.1007/s11010-015-2396-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 03/27/2015] [Indexed: 01/26/2023]
Abstract
The proinflammatory factor high mobility group box protein 1 (HMGB1) has been implicated as an important mediator of many chronic inflammatory diseases, including asthma. Human bronchial epithelial cells (HBECs) play a central role in the pathogenesis of asthma. However, the effects of HMGB1 on HBECs and the underlying mechanisms remain unknown. Here, we investigated receptor expression and proinflammatory cytokine production by primary cultures of HBECs stimulated by HMGB1. We then examined the effects of specific receptor blockade and inhibition of p38 MAPK, ERK1/2, or PI3-K on HMGB1-induced expression of proinflammatory cytokines. HMGB1 increased the expression and secretion of TNF-α, TSLP, MMP-9, and VEGF in a dose- and time-dependent manner. HMGB1 also induced elevated expression of RAGE protein. Secretion of TNF-α, VEGF, MMP-9, and TSLP was significantly decreased by RAGE blockade and p38 MAPK pathway inhibition, while a less pronounced effect was mediated by ERK1/2 inhibition. These observations suggest that HMGB1 binds RAGE and promotes activities of p38 MAPK and ERK1/2 pathways in HBECs. This then enhances the expression of TNF-α, VEGF, MMP-9, and TSLP, which are the important inflammatory factors in asthma. These results demonstrate that HMGB1 enhances the inflammatory responses of HBECs, which are involved in the modulation of inflammatory processes in asthma.
Collapse
Affiliation(s)
- Yue Liang
- The Research Department of Respiration Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China,
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Nakamura Y, Suzuki S, Shimizu T, Miyata M, Shishido T, Ikeda K, Saitoh SI, Kubota I, Takeishi Y. High Mobility Group Box 1 Promotes Angiogenesis from Bone Marrow-derived Endothelial Progenitor Cells after Myocardial Infarction. J Atheroscler Thromb 2015; 22:570-81. [PMID: 25735431 DOI: 10.5551/jat.27235] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
AIMS High mobility group box 1 (HMGB1) is a DNA-binding protein secreted into the extracellular space from necrotic cells that acts as a cytokine. We examined the role of HMGB1 in angiogenesis from bone marrow-derived cells in the heart using transgenic mice exhibiting the cardiac-specific overexpression of HMGB1 (HMGB1-TG). METHODS HMGB1-TG mice and wild-type littermate (WT) mice were lethally irradiated and injected with bone marrow cells from green fluorescent protein mice through the tail vein. After bone marrow transplantation, the left anterior descending artery was ligated to induce myocardial infarction (MI). RESULTS Flow cytometry revealed that the levels of circulating endothelial progenitor cells (EPCs) mobilized from the bone marrow increased after MI in the HMGB-TG mice versus the WT mice. In addition, the size of MI was smaller in the HMGB1-TG mice than in the WT mice, and immunofluorescence staining demonstrated that the number of engrafted vascular endothelial cells derived from bone marrow in the border zones of the MI areas was increased in the HMGB1-TG mice compared to that observed in the WT mice. Moreover, the levels of cardiac vascular endothelial growth factor after MI were higher in the HMGB1-TG mice than in the WT mice. CONCLUSIONS The present study demonstrated that HMGB1 promotes angiogenesis and reduces the MI size by enhancing the mobilization and differentiation of bone marrow cells to EPCs as well as their migration to the border zones of the MI areas and engraftment as vascular endothelial cells in new capillaries or arterioles in the infarcted heart.
Collapse
Affiliation(s)
- Yuichi Nakamura
- Department of Cardiology and Hematology, Fukushima Medical University
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Inoue S, Kiriyama K, Hatanaka Y, Kanoh H. Adsorption properties of an activated carbon for 18 cytokines and HMGB1 from inflammatory model plasma. Colloids Surf B Biointerfaces 2014; 126:58-62. [PMID: 25543984 DOI: 10.1016/j.colsurfb.2014.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/07/2014] [Indexed: 10/24/2022]
Abstract
The ability of an activated carbon (AC) to adsorb 18 different cytokines with molecular weights ranging from 8 kDa to 70 kDa and high mobility group box-1 (HMGB1) from inflammatory model plasma at 310 K and the mechanisms of adsorption were examined. Porosity analysis using N2 gas adsorption at 77K showed that the AC had micropores with diameters of 1-2 nm and mesopores with diameters of 5-20 nm. All 18 cytokines and HMGB1 were adsorbed on the AC; however, the shapes of the adsorption isotherms changed depending on the molecular weight. The adsorption isotherms for molecules of 8-10 kDa, 10-20 kDa, 20-30 kDa, and higher molecular weights were classified as H-2, L-3, S-3, and S-1 types, respectively. These results suggested that the adsorption mechanism for the cytokines and HMGB1 in the mesopores and on the surface of the AC differed as a function of the molecular weight. On the basis of these results, it can be concluded that AC should be efficient for cytokine adsorption.
Collapse
Affiliation(s)
- Satoru Inoue
- Medical Material Laboratory, Asahi-Kasei Medical Co., Ltd., 2111-2, Oaza-sato, Oita 870-0396, Japan; Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan
| | - Kentaro Kiriyama
- Medical Material Laboratory, Asahi-Kasei Medical Co., Ltd., 2111-2, Oaza-sato, Oita 870-0396, Japan
| | - Yoshihiro Hatanaka
- Medical Material Laboratory, Asahi-Kasei Medical Co., Ltd., 2111-2, Oaza-sato, Oita 870-0396, Japan
| | - Hirofumi Kanoh
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage, Chiba 263-8522, Japan.
| |
Collapse
|
48
|
|
49
|
Momonaka H, Hasegawa S, Matsushige T, Inoue H, Kajimoto M, Okada S, Nakatsuka K, Morishima T, Ichiyama T. High mobility group box 1 in patients with 2009 pandemic H1N1 influenza-associated encephalopathy. Brain Dev 2014; 36:484-8. [PMID: 23907181 DOI: 10.1016/j.braindev.2013.07.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 06/20/2013] [Accepted: 07/04/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Patients with 2009 pandemic H1N1 influenza-associated encephalopathy (pIE) have been reported in Japan. The most common clinical symptoms of this condition are seizures and progressive coma with high-grade fever. We previously highlighted the cytokine profile of pIE; our results suggest that proinflammatory cytokines play an important role in the pathogenesis. High mobility group box 1 (HMGB1) protein is a late mediator of inflammation or sepsis. However, there are few reports regarding the serum and cerebrospinal fluid (CSF) levels of HMGB1 in pIE patients. METHODS We measured serum and CSF levels of HMGB1 in the following: pIE patients with poor outcomes, pIE patients without neurological sequelae, influenza patients without pIE, and control subjects. RESULTS Serum HMGB1 levels were significantly higher in pIE patients with poor outcomes compared to those without neurological sequelae. In contrast, there was no difference in CSF HMGB1 levels among all groups. Regarding pIE patients, we found a significant positive correlation between HMGB1 levels and IL-6 in the serum but not in the CSF. CONCLUSIONS Our results suggest that HMGB1 protein may be involved in the pathogenesis of pIE and that a high serum, but not CSF, level of inflammatory cytokines plays an important role in the severity of pIE.
Collapse
Affiliation(s)
- Hiroshi Momonaka
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan.
| | - Takeshi Matsushige
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Hirofumi Inoue
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Madoka Kajimoto
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | - Seigo Okada
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| | | | - Tsuneo Morishima
- Department of Pediatrics, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan
| | - Takashi Ichiyama
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
50
|
XXXIV International Congress of the European Association of Poisons Centres and Clinical Toxicologists (EAPCCT) 27–30 May 2014, Brussels, Belgium. Clin Toxicol (Phila) 2014. [DOI: 10.3109/15563650.2014.906213] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|