251
|
Key role for neutrophils in radiation-induced antitumor immune responses: Potentiation with G-CSF. Proc Natl Acad Sci U S A 2016; 113:11300-11305. [PMID: 27651484 DOI: 10.1073/pnas.1613187113] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Radiation therapy (RT), a major modality for treating localized tumors, can induce tumor regression outside the radiation field through an abscopal effect that is thought to involve the immune system. Our studies were designed to understand the early immunological effects of RT in the tumor microenvironment using several syngeneic mouse tumor models. We observed that RT induced sterile inflammation with a rapid and transient infiltration of CD11b+Gr-1high+ neutrophils into the tumors. RT-recruited tumor-associated neutrophils (RT-Ns) exhibited an increased production of reactive oxygen species and induced apoptosis of tumor cells. Tumor infiltration of RT-Ns resulted in sterile inflammation and, eventually, the activation of tumor-specific cytotoxic T cells, their recruitment into the tumor site, and tumor regression. Finally, the concurrent administration of granulocyte colony-stimulating factor (G-CSF) enhanced RT-mediated antitumor activity by activating RT-Ns. Our results suggest that the combination of RT and G-CSF should be further evaluated in preclinical and clinical settings.
Collapse
|
252
|
Caligiuri A, Gentilini A, Marra F. Molecular Pathogenesis of NASH. Int J Mol Sci 2016; 17:ijms17091575. [PMID: 27657051 PMCID: PMC5037841 DOI: 10.3390/ijms17091575] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is the main cause of chronic liver disease in the Western world and a major health problem, owing to its close association with obesity, diabetes, and the metabolic syndrome. NASH progression results from numerous events originating within the liver, as well as from signals derived from the adipose tissue and the gastrointestinal tract. In a fraction of NASH patients, disease may progress, eventually leading to advanced fibrosis, cirrhosis and hepatocellular carcinoma. Understanding the mechanisms leading to NASH and its evolution to cirrhosis is critical to identifying effective approaches for the treatment of this condition. In this review, we focus on some of the most recent data reported on the pathogenesis of NASH and its fibrogenic progression, highlighting potential targets for treatment or identification of biomarkers of disease progression.
Collapse
Affiliation(s)
- Alessandra Caligiuri
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Alessandra Gentilini
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| | - Fabio Marra
- Dipartimento di Medicina Sperimentale e Clinica, Università degli Studi di Firenze, Firenze 50121, Italy.
| |
Collapse
|
253
|
Tian Y, Pan D, Chordia MD, French BA, Kron IL, Yang Z. The spleen contributes importantly to myocardial infarct exacerbation during post-ischemic reperfusion in mice via signaling between cardiac HMGB1 and splenic RAGE. Basic Res Cardiol 2016; 111:62. [PMID: 27645145 DOI: 10.1007/s00395-016-0583-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
Abstract
The spleen plays a critical role in post-infarct myocardial remodeling. However, the role of the spleen in exacerbating myocardial infarction (MI) during acute ischemia/reperfusion (I/R) injury is unknown. The present study tests the hypothesis that splenic leukocytes are activated by substances released from ischemic myocardium to subsequently exacerbate myocardial injury during reperfusion. The left coronary artery in C57BL/6 mice underwent various durations of occlusion followed by 60 min of reperfusion (denoted as min/min of I/R) with or without splenectomy prior to I/R injury. Splenectomy significantly decreased myocardial infarct size (IS) in 40'/60' and 50'/60' groups (p < 0.05); however, it had no effect on IS in 10'/60', 20'/60' and 30'/60' groups (p = NS). In the 20'/60' group, infusion of 40-min ischemic heart homogenate (40-IHH) upon reperfusion increased IS by >threefold versus infusion of 10-IHH (p < 0.05). Splenectomy abolished the infarct-exacerbating effect of 40-IHH, which was restored by splenic leukocyte adoptive transfer (SPAT). Furthermore, depletion of HMGB1 in the 40-IHH group abolished its infarct-exacerbating effect (p < 0.05), and 40-IHH failed to increase IS in both RAGE(-/-) mice and splenectomized wild-type mice with SPAT from RAGE(-/-) mice. The injection of 40-IHH significantly increased formyl peptide receptor 1 (FPR1) expression in sham spleens when compared to 10-IHH-treated sham and control mice. cFLFLF, a specific FPR1 antagonist, reduced myocardial neutrophil infiltration and abrogated the infarct-exacerbating effect of 40-IHH during reperfusion. A cardio (HMGB1)-splenic (RAGE receptor) signaling axis exists and contributes to myocardial infarct exacerbation during reperfusion after prolonged ischemic insults by activating splenic leukocytes. The FPR1 is a potential therapeutic target for inhibiting the cardio-splenic axis that augments infarct size during post-ischemic reperfusion.
Collapse
Affiliation(s)
- Yikui Tian
- Department of Cardiovascular Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Surgery, University of Virginia, P.O. Box 800709, Charlottesville, VA, 22908, USA
| | - Dongfeng Pan
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | - Mahendra D Chordia
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | - Brent A French
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Irving L Kron
- Department of Surgery, University of Virginia, P.O. Box 800709, Charlottesville, VA, 22908, USA
| | - Zequan Yang
- Department of Surgery, University of Virginia, P.O. Box 800709, Charlottesville, VA, 22908, USA.
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
254
|
Regulation of tissue infiltration by neutrophils: role of integrin α3β1 and other factors. Curr Opin Hematol 2016; 23:36-43. [PMID: 26554893 DOI: 10.1097/moh.0000000000000198] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Neutrophils have traditionally been viewed in the context of acute infection and inflammation forming the first line of defense against invading pathogens. Neutrophil trafficking to the site of inflammation requires adhesion and transmigration through blood vessels, which is orchestrated by adhesion molecules, such as β2 and β1-integrins, chemokines, and cytokines. The review focuses on recent advances in understanding the regulators of neutrophil recruitment during inflammation in both acute and chronic settings. RECENT FINDINGS Recent findings suggest that besides the established pathways of selectin or chemokine-mediated integrin activation, signaling by distinct Toll-like receptors (TLRs) (especially TLR2, TLR4, and TLR5) can activate integrin-dependent neutrophil adhesion. Moreover, the integrin α3β1 has been vitally implicated as a new player in neutrophil recruitment and TLR-mediated responses in septic inflammation. Furthermore, several endogenous inhibitory mechanisms of leukocyte recruitment have been identified, including the secreted molecules Del-1, PTX3, and GDF-15, which block distinct steps of the leukocyte adhesion cascade, as well as novel regulatory signaling pathways, involving the protein kinase AKT1 and IFN-λ2/IL-28A. SUMMARY The leukocyte adhesion cascade is a tightly regulated process, subjected to both positive and negative regulators. Dysregulation of this process and hence neutrophil recruitment can lead to the development of inflammatory and autoimmune diseases.
Collapse
|
255
|
Disulfide HMGB1 derived from platelets coordinates venous thrombosis in mice. Blood 2016; 128:2435-2449. [PMID: 27574188 DOI: 10.1182/blood-2016-04-710632] [Citation(s) in RCA: 218] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 08/12/2016] [Indexed: 01/12/2023] Open
Abstract
Deep venous thrombosis (DVT) is one of the most common cardiovascular diseases, but its pathophysiology remains incompletely understood. Although sterile inflammation has recently been shown to boost coagulation during DVT, the underlying molecular mechanisms are not fully resolved, which could potentially identify new anti-inflammatory approaches to prophylaxis and therapy of DVT. Using a mouse model of venous thrombosis induced by flow reduction in the vena cava inferior, we identified blood-derived high-mobility group box 1 protein (HMGB1), a prototypical mediator of sterile inflammation, to be a master regulator of the prothrombotic cascade involving platelets and myeloid leukocytes fostering occlusive DVT formation. Transfer of platelets into Hmgb1-/- chimeras showed that this cell type is the major source of HMGB1, exposing reduced HMGB1 on their surface upon activation thereby enhancing the recruitment of monocytes. Activated leukocytes in turn support oxidation of HMGB1 unleashing its prothrombotic activity and promoting platelet aggregation. This potentiates the amount of HMGB1 and further nurtures the accumulation and activation of monocytes through receptor for advanced glycation end products (RAGE) and Toll-like receptor 2, leading to local delivery of monocyte-derived tissue factor and cytokines. Moreover, disulfide HMGB1 facilitates formation of prothrombotic neutrophil extracellular traps (NETs) mediated by RAGE, exposing additional HMGB1 on their extracellular DNA strands. Eventually, a vicious circle of coagulation and inflammation is set in motion leading to obstructive DVT formation. Therefore, platelet-derived disulfide HMGB1 is a central mediator of the sterile inflammatory process in venous thrombosis and could be an attractive target for an anti-inflammatory approach for DVT prophylaxis.
Collapse
|
256
|
Reed KR, Song F, Young MA, Hassan N, Antoine DJ, Gemici NPB, Clarke AR, Jenkins JR. Secreted HMGB1 from Wnt activated intestinal cells is required to maintain a crypt progenitor phenotype. Oncotarget 2016; 7:51665-51673. [PMID: 27323825 PMCID: PMC5239505 DOI: 10.18632/oncotarget.10076] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/29/2016] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND AIMS Colorectal cancer (CRC) arises via multiple genetic changes. Mutation of the tumour suppressor gene APC, a key regulator of Wnt signalling, is recognised as a frequent early driving mutation in CRC. We have previously shown that conditional loss of Apc within the murine small intestine (Apcfloxmice) results in acute Wnt signalling activation, altered crypt-villus architecture and many hallmarks of neoplasia. Our transctipomic profiling (Affymetrix Microarrays) and proteomic profiling (iTRAQ-QSTAR) of Apc-deficient intestine inferred the involvement of High Mobility Group Box 1 (Hmgb1) in CRC pathogenesis. Here we assess the contribution of HMGB1 to the crypt progenitor phenotype seen following Apc loss. RESULTS Elevated HMGB1 was confirmed in intestinal epithelia and serum following conditional loss of Apc. Treatment of Apcflox mice with anti-HMGB1 neutralising antibody significantly reduced many of the crypt progenitor phenotypes associated with Apc loss; proliferation and apoptosis levels were reduced, cell differentiation was restored and the expansion of stem cell marker expression was eradicated. METHODS Hmgb1 levels in intestinal epithelia and serum in Apcflox and ApcMin mice were assessed using qRT-PCR, Western blot and ELISA assays. The functional importance of elevated extracellular Hmgb1 was assessed using an anti-HMGB1 neutralising antibody in Apcflox mice. CONCLUSIONS HMGB1 is expressed and secreted from intestinal epithelial cells in response to Wnt signalling activation. This secreted HMGB1 is required to maintain nearly all aspects of the crypt progenitor phenotype observed following Apc loss and add to the body of accumulating evidence indicating that targeting HMGB1 may be a viable novel therapeutic approach.
Collapse
Affiliation(s)
- Karen R. Reed
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Fei Song
- Infrafrontier GmbH, Neuherberg / München, 85764, Germany
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Maddy A. Young
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Nurudeen Hassan
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- Cardiff School of Health Sciences at Cardiff Metropolitan University, Cardiff, CF5 2YB, UK
| | - Daniel J. Antoine
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| | - Nesibe-Princess B. Gemici
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Alan R. Clarke
- European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - John R. Jenkins
- Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK
| |
Collapse
|
257
|
Affiliation(s)
- Peter Huebener
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | |
Collapse
|
258
|
Wang HJ, Murray GJ, Jung MK. Host homeostatic responses to alcohol-induced cellular stress in animal models of alcoholic liver disease. Expert Rev Gastroenterol Hepatol 2016; 9:1193-205. [PMID: 26293978 DOI: 10.1586/17474124.2015.1069705] [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] [Indexed: 11/08/2022]
Abstract
Humans develop various clinical phenotypes of severe alcoholic liver disease, including alcoholic hepatitis and cirrhosis, generally after decades of heavy drinking. In such individuals, following each episode of drinking, their livers experience heightened intracellular and extracellular stresses that are closely associated with alcohol consumption and alcohol metabolism. This article focuses on the latest advances made in animal models on evolutionarily conserved homeostatic mechanisms for coping with and resolving these stress conditions. The mechanisms discussed include the stress-activated protein kinase JNK, energy regulator AMPK, autophagy and the inflammatory response. Over time, the host may respond variably to stress with protective mechanisms that are critical in determining an individual's vulnerability to developing severe alcoholic liver disease. A systematic review of these mechanisms and their temporal changes in animal models provides the basis for general conclusions, and raises questions for future studies. The relevance of these data to human conditions is also discussed.
Collapse
Affiliation(s)
- He Joe Wang
- a Division of Metabolism and Health Effect, National Institute of Alcohol Abuse and Alcoholism/NIH, 5635 Fishers Lane, MSC 9304, Bethesda, MD 20892-9304, USA
| | | | | |
Collapse
|
259
|
Mühl H. STAT3, a Key Parameter of Cytokine-Driven Tissue Protection during Sterile Inflammation - the Case of Experimental Acetaminophen (Paracetamol)-Induced Liver Damage. Front Immunol 2016; 7:163. [PMID: 27199988 PMCID: PMC4852172 DOI: 10.3389/fimmu.2016.00163] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 04/15/2016] [Indexed: 12/12/2022] Open
Abstract
Acetaminophen (APAP, N-acetyl-p-aminophenol, or paracetamol) overdosing is a prevalent cause of acute liver injury. While clinical disease is initiated by overt parenchymal hepatocyte necrosis in response to the analgetic, course of intoxication is substantially influenced by associated activation of innate immunity. This process is supposed to be set in motion by release of danger-associated molecular patterns (DAMPs) from dying hepatocytes and is accompanied by an inflammatory cytokine response. Murine models of APAP-induced liver injury emphasize the complex role that DAMPs and cytokines play in promoting either hepatic pathogenesis or resolution and recovery from intoxication. Whereas the function of key inflammatory cytokines is controversially discussed, a subclass of specific cytokines capable of efficiently activating the hepatocyte signal transducer and activator of transcription (STAT)-3 pathway stands out as being consistently protective in murine models of APAP intoxication. Those include foremost interleukin (IL)-6, IL-11, IL-13, and IL-22. Above all, activation of STAT3 under the influence of these cytokines has the capability to drive hepatocyte compensatory proliferation, a key principle of the regenerating liver. Herein, the role of these specific cytokines during experimental APAP-induced liver injury is highlighted and discussed in a broader perspective. In hard-to-treat or at-risk patients, standard therapy may fail and APAP intoxication can proceed toward a fatal condition. Focused administration of recombinant STAT3-activating cytokines may evolve as novel therapeutic approach under those ill-fated conditions.
Collapse
Affiliation(s)
- Heiko Mühl
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt am Main , Frankfurt am Main , Germany
| |
Collapse
|
260
|
Arrese M, Cabrera D, Kalergis AM, Feldstein AE. Innate Immunity and Inflammation in NAFLD/NASH. Dig Dis Sci 2016; 61:1294-303. [PMID: 26841783 PMCID: PMC4948286 DOI: 10.1007/s10620-016-4049-x] [Citation(s) in RCA: 368] [Impact Index Per Article: 40.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 01/19/2016] [Indexed: 02/06/2023]
Abstract
Inflammation and hepatocyte injury and death are the hallmarks of nonalcoholic steatohepatitis (NASH), the progressive form of nonalcoholic fatty liver disease (NAFLD), which is a currently burgeoning public health problem. Innate immune activation is a key factor in triggering and amplifying hepatic inflammation in NAFLD/NASH. Thus, identification of the underlying mechanisms by which immune cells in the liver recognize cell damage signals or the presence of pathogens or pathogen-derived factors that activate them is relevant from a therapeutic perspective. In this review, we present new insights into the factors promoting the inflammatory response in NASH including sterile cell death processes resulting from lipotoxicity in hepatocytes as well as into the altered gut-liver axis function, which involves translocation of bacterial products into portal circulation as a result of gut leakiness. We further delineate the key immune cell types involved and how they recognize both damage-associated molecular patterns or pathogen-associated molecular patterns through binding of surface-expressed pattern recognition receptors, which initiate signaling cascades leading to injury amplification. The relevance of modulating these inflammatory signaling pathways as potential novel therapeutic strategies for the treatment of NASH is summarized.
Collapse
Affiliation(s)
- Marco Arrese
- Departmento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Departmento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Ciencias Químicas y Biológicas, Facultad de Salud, Universidad Bernardo O Higgins, Santiago, Chile
| | - Alexis M Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ariel E Feldstein
- Department of Pediatrics, University of California San Diego (UCSD), San Diego, CA, USA.
- Rady Children's Hospital, San Diego, CA, USA.
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, UCSD, 3020 Children's Way, MC 5030, San Diego, CA, 92103-8450, USA.
| |
Collapse
|
261
|
Boyapati RK, Rossi AG, Satsangi J, Ho GT. Gut mucosal DAMPs in IBD: from mechanisms to therapeutic implications. Mucosal Immunol 2016; 9:567-82. [PMID: 26931062 DOI: 10.1038/mi.2016.14] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 12/31/2015] [Indexed: 02/06/2023]
Abstract
Endogenous damage-associated molecular patterns (DAMPs) are released during tissue damage and have increasingly recognized roles in the etiology of many human diseases. The inflammatory bowel diseases (IBD), ulcerative colitis (UC) and Crohn's disease (CD), are immune-mediated conditions where high levels of DAMPs are observed. DAMPs such as calprotectin (S100A8/9) have an established clinical role as a biomarker in IBD. In this review, we use IBD as an archetypal common chronic inflammatory disease to focus on the conceptual and evidential importance of DAMPs in pathogenesis and why DAMPs represent an entirely new class of targets for clinical translation.
Collapse
Affiliation(s)
- R K Boyapati
- MRC Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, UK.,Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - A G Rossi
- MRC Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, UK
| | - J Satsangi
- Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - G-T Ho
- MRC Centre for Inflammation Research, Queens Medical Research Institute, Edinburgh, UK.,Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
262
|
Abstract
Evidence suggests that light and circadian rhythms profoundly influence the physiologic capacity with which an organism responds to stress. However, the ramifications of light spectrum on the course of critical illness remain to be determined. Here, we show that acute exposure to bright blue spectrum light reduces organ injury by comparison with bright red spectrum or ambient white fluorescent light in two murine models of sterile insult: warm liver ischemia/reperfusion (I/R) and unilateral renal I/R. Exposure to bright blue light before I/R reduced hepatocellular injury and necrosis and reduced acute kidney injury and necrosis. In both models, blue light reduced neutrophil influx, as evidenced by reduced myeloperoxidase (MPO) within each organ, and reduced the release of high-mobility group box 1 (HMGB1), a neutrophil chemotactant and key mediator in the pathogenesis of I/R injury. The protective mechanism appeared to involve an optic pathway and was mediated, in part, by a sympathetic (β3 adrenergic) pathway that functioned independent of significant alterations in melatonin or corticosterone concentrations to regulate neutrophil recruitment. These data suggest that modifying the spectrum of light may offer therapeutic utility in sterile forms of cellular injury.
Collapse
|
263
|
Lea JD, Clarke JI, McGuire N, Antoine DJ. Redox-Dependent HMGB1 Isoforms as Pivotal Co-Ordinators of Drug-Induced Liver Injury: Mechanistic Biomarkers and Therapeutic Targets. Antioxid Redox Signal 2016; 24:652-65. [PMID: 26481429 DOI: 10.1089/ars.2015.6406] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
SIGNIFICANCE High-mobility group box 1 (HMGB1) is a critical protein in the coordination of the inflammatory response in drug-induced liver injury (DILI). HMGB1 is released from necrotic hepatocytes and activated immune cells. The extracellular function of HMGB1 is dependent upon redox modification of cysteine residues that control chemoattractant and cytokine-inducing properties. Existing biomarkers of DILI such as alanine aminotransferase (ALT) have limitations such as lack of sensitivity and tissue specificity that can adversely affect clinical intervention. RECENT ADVANCES HMGB1 isoforms have been shown to be more sensitive biomarkers than ALT for predicting DILI development and the requirement for liver transplant following acetaminophen (APAP) overdose. Hepatocyte-specific conditional knockout of HMGB1 has demonstrated the pivotal role of HMGB1 in DILI and liver disease. Tandem mass spectrometry (MS/MS) enables the characterization and quantification of different mechanism-dependent post-translationally modified isoforms of HMGB1. CRITICAL ISSUES HMGB1 shows great promise as a biomarker of DILI. However, current diagnostic assays are either too time-consuming to be clinically applicable (MS/MS) or are unable to distinguish between different redox and acetyl isoforms of HMGB1 (ELISA). Additionally, HMGB1 is not liver specific, so while it outperforms ALT (also not liver specific) as a biomarker for the prediction of DILI development, it should be used in a biomarker panel along with liver-specific markers such as miR-122. FUTURE DIRECTIONS A point-of-care test for HMGB1 and the development of redox and acetyl isoform-targeting antibodies will advance clinical utility. Work is ongoing to validate baseline levels of circulating HMGB1 in healthy volunteers.
Collapse
Affiliation(s)
- Jonathan D Lea
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool , Liverpool, United Kingdom
| | - Joanna I Clarke
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool , Liverpool, United Kingdom
| | - Niamh McGuire
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool , Liverpool, United Kingdom
| | - Daniel J Antoine
- Department of Molecular and Clinical Pharmacology, MRC Centre for Drug Safety Science, University of Liverpool , Liverpool, United Kingdom
| |
Collapse
|
264
|
Damage-associated molecular patterns in cancer: a double-edged sword. Oncogene 2016; 35:5931-5941. [PMID: 27086930 PMCID: PMC5119456 DOI: 10.1038/onc.2016.104] [Citation(s) in RCA: 338] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 12/14/2022]
Abstract
Damage-associated molecular patterns (DAMPs) are released in response to cell
death and stress, and are potent triggers of sterile inflammation. Recent evidence
suggests that DAMPs may also have a key role in the development of cancer as well as in
the host response to cytotoxic anti-tumor therapy. As such, DAMPs may exert protective
functions by alerting the immune system to the presence of dying tumor cells, thereby
triggering immunogenic tumor cell death. On the other hand, cell death and release of
DAMPs may also trigger chronic inflammation and thereby promote the development or
progression of tumors. Here, we will review the contribution of candidate DAMPs and their
receptors and discuss the evidence for DAMPs as tumor-promoting and anti-tumor effectors
as well as unsolved questions such as DAMP release from non-tumor cells as well as the
existence of tumor-specific DAMPs.
Collapse
|
265
|
Fu GX, Chen AF, Zhong Y, Zhao J, Gu YJ. Decreased serum level of HMGB1 and MyD88 during human aging progress in healthy individuals. Aging Clin Exp Res 2016; 28:175-80. [PMID: 26130428 DOI: 10.1007/s40520-015-0402-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 06/12/2015] [Indexed: 12/15/2022]
Abstract
AIMS Previous studies have suggested that high mobility group box-1 protein (HMGB1) binds to the toll-like receptor 4 (TLR4) signaling mediates the progression of various inflammatory diseases. But the roles of HMGB1 and TLR4 in aging remain poorly unknown. In this study, we aimed to investigate the serum levels of HMGB1 and myeloid differentiation factor 88 (MyD88), which is one of TLR4's intracellular adaptor proteins during human aging process and their relevance with cathepsin B (CTSB). METHODS This research was conducted using the blood samples provided by healthy people (n = 90, 63 men and 27 women). Subjects were subdivided into groups with respect to age: young (about 25 years old, n = 30), middle age (about 40 years old, n = 30), and aged (above 65 years old, n = 30). Altered serum levels of HMGB1, MyD88 and CTSB were measured using an enzyme-linked immunosorbent assay. RESULTS The serum levels of HMGB1 and MyD88 were significantly decreased in the aged group compared with those in the young group. Linear regression analysis showed that HMGB1 and MyD88 positively correlated with CTSB among the whole healthy people. A negative correlation was determined between MyD88 and age. CONCLUSIONS The serum levels of HMGB1 and MyD88 significantly decreased with age. MyD88, but not HMGB1, was negatively correlated with age.
Collapse
Affiliation(s)
- Guo-Xiang Fu
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan road, Shanghai, 200233, People's Republic of China.
| | - Alex F Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Yuan Zhong
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600, Yi Shan road, Shanghai, 200233, People's Republic of China
| | - Jian Zhao
- Shanghai Sixth People's Hospital Jinshan Branch, No. 147 Health Road, Zhujing Town, Jinshan District, Shanghai, 201500, People's Republic of China
| | - Ying-Jia Gu
- Shanghai Sixth People's Hospital Jinshan Branch, No. 147 Health Road, Zhujing Town, Jinshan District, Shanghai, 201500, People's Republic of China
| |
Collapse
|
266
|
Thieblemont N, Wright HL, Edwards SW, Witko-Sarsat V. Human neutrophils in auto-immunity. Semin Immunol 2016; 28:159-73. [DOI: 10.1016/j.smim.2016.03.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 03/08/2016] [Accepted: 03/12/2016] [Indexed: 01/06/2023]
|
267
|
Furuta K, Yoshida Y, Ogura S, Kurahashi T, Kizu T, Maeda S, Egawa M, Chatani N, Nishida K, Nakaoka Y, Kiso S, Kamada Y, Takehara T. Gab1 adaptor protein acts as a gatekeeper to balance hepatocyte death and proliferation during acetaminophen-induced liver injury in mice. Hepatology 2016; 63:1340-55. [PMID: 26680679 DOI: 10.1002/hep.28410] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Accepted: 12/11/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Acetaminophen (APAP) overdose is the leading cause of drug-induced acute liver failure. In APAP-induced acute liver failure, hepatocyte death and subsequent liver regeneration determines the prognosis of patients, making it necessary to identify suitable therapeutic targets based on detailed molecular mechanisms. Grb2-associated binder 1 (Gab1) adaptor protein plays a crucial role in transmitting signals from growth factor and cytokine receptors to downstream effectors. In this study, we hypothesized that Gab1 is involved in APAP-induced acute liver failure. Hepatocyte-specific Gab1 conditional knockout (Gab1CKO) and control mice were treated with 250 mg/kg of APAP. After APAP treatment, Gab1CKO mice had significantly higher mortality and elevated serum alanine aminotransferase levels compared to control mice. Gab1CKO mice had increased hepatocyte death and increased serum levels of high mobility group box 1, a marker of hepatocyte necrosis. In addition, Gab1CKO mice had reduced hepatocyte proliferation. The enhanced hepatotoxicity in Gab1CKO mice was associated with increased activation of stress-related c-Jun N-terminal kinase (JNK) and reduced activation of extracellular signal-regulated kinase and AKT. Furthermore, Gab1CKO mice showed enhanced mitochondrial translocation of JNK accompanied by an increase in the release of mitochondrial enzymes into the cytosol, which is indicative of increased mitochondrial dysfunction and subsequent nuclear DNA fragmentation. Finally, in vitro experiments showed that Gab1-deficient hepatocytes were more susceptible to APAP-induced mitochondrial dysfunction and cell death, suggesting that hepatocyte Gab1 is a direct target of APAP-induced hepatotoxicity. CONCLUSION Our current data demonstrate that hepatocyte Gab1 plays a critical role in controlling the balance between hepatocyte death and compensatory hepatocyte proliferation during APAP-induced liver injury.
Collapse
Affiliation(s)
- Kunimaro Furuta
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuichi Yoshida
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Satoshi Ogura
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Tomohide Kurahashi
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Takashi Kizu
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinichiro Maeda
- Department of Pharmacy, Osaka University Hospital, Suita, Osaka, Japan
| | - Mayumi Egawa
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Norihiro Chatani
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Keigo Nishida
- Laboratory of Immune Regulation, Faculty of Pharmaceutical Sciences, Suzuka University of Medical Science, Suzuka, Mie, Japan
| | - Yoshikazu Nakaoka
- Department of Cardiovascular Medicine, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO), Japan Science Technology Agency, Kawaguchi, Saitama, Japan
| | - Shinichi Kiso
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Yoshihiro Kamada
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan.,Departments of Molecular Biochemistry and Clinical Investigation, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
268
|
Clarke JI, Dear JW, Antoine DJ. Recent advances in biomarkers and therapeutic interventions for hepatic drug safety – false dawn or new horizon? Expert Opin Drug Saf 2016; 15:625-34. [DOI: 10.1517/14740338.2016.1160057] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Joanna I. Clarke
- MRC Centre for Drug Safety Science and Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - James W. Dear
- Pharmacology, Toxicology and Therapeutics Unit, BHF/University Centre for Cardiovascular Science, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Daniel J. Antoine
- MRC Centre for Drug Safety Science and Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
269
|
Xia P, Deng Q, Gao J, Yu X, Zhang Y, Li J, Guan W, Hu J, Tan Q, Zhou L, Han W, Yuan Y, Yu Y. Therapeutic effects of antigen affinity-purified polyclonal anti-receptor of advanced glycation end-product (RAGE) antibodies on cholestasis-induced liver injury in rats. Eur J Pharmacol 2016; 779:102-10. [PMID: 26970185 DOI: 10.1016/j.ejphar.2016.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 01/22/2023]
Abstract
Cholestasis leads to acute hepatic injury, fibrosis/cirrhosis, inflammation, and duct proliferation. We investigated whether blocking receptor of advanced glycation end-products (RAGE) with polyclonal anti-RAGE antibodies (anti-RAGE) could regulate acute liver injury and fibrosis in a rat bile duct ligation (BDL) model. Male Wister rats received 0.5mg/kg rabbit anti-RAGE or an equal amount of rabbit IgG by subcutaneous injection twice a week after BDL. Samples of liver tissue and peripheral blood were collected at 14 days after BDL. Serum biochemistry and histology were used to analyze the degree of liver injury. Quantitative real-time PCR (qPCR) and immunohistochemical staining were used to further analyze liver injury. Anti-RAGE improved the gross appearance of the liver and the rat survival rate. Liver tissue histology and relevant serum biochemistry indicated that anti-RAGE attenuated liver necrosis, inflammation, liver fibrosis, and duct proliferation in the BDL model. qPCR and western blotting showed significant reductions in interleukin-1β expression levels in the liver by treatment with anti-RAGE. Anti-RAGE also significantly reduced the mRNA levels of α1(1) collagen (Col1α1) and cholesterol 7α-hydroxylase, and the ratio of tissue inhibitor of matrix metalloproteinase-1 to matrix metalloproteinases (MMPs) in the liver. In addition, anti-RAGE regulated the transcriptional level of Col1α1 and MMP-9 in transforming growth factor-β-induced activated LX-2 cells in vitro. Anti-RAGE was found to inhibit hepatic stellate cell proliferation in vivo and in vitro. Therefore, anti-RAGE can protect the liver from injury induced by BDL in rats.
Collapse
Affiliation(s)
- Peng Xia
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Qing Deng
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jin Gao
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Xiaolan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Yang Zhang
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jingjing Li
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Wen Guan
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Jianjun Hu
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd., Xuhui, Shanghai 200233, China
| | - Quanhui Tan
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yishan Rd., Xuhui, Shanghai 200233, China
| | - Liang Zhou
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Wei Han
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China
| | - Yunsheng Yuan
- Laboratory of Regenerative Medicine, School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China; Engineering Research Center of Cell and Therapeutic Antibody, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China.
| | - Yan Yu
- Shanghai Municipality Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang, Shanghai 200240, China.
| |
Collapse
|
270
|
Holman NS, Mosedale M, Wolf KK, LeCluyse EL, Watkins PB. Subtoxic Alterations in Hepatocyte-Derived Exosomes: An Early Step in Drug-Induced Liver Injury? Toxicol Sci 2016; 151:365-75. [PMID: 26962055 DOI: 10.1093/toxsci/kfw047] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Drug-induced liver injury (DILI) is a significant clinical and economic problem in the United States, yet the mechanisms that underlie DILI remain poorly understood. Recent evidence suggests that signaling molecules released by stressed hepatocytes can trigger immune responses that may be common across DILI mechanisms. Extracellular vesicles released by hepatocytes, principally hepatocyte-derived exosomes (HDEs), may constitute one such signal. To examine HDE alterations as a function of drug-induced stress, this work utilized prototypical hepatotoxicant acetaminophen (APAP) in male Sprague-Dawley (SD) rats, SD rat hepatocytes, and primary human hepatocytes. HDE were isolated using ExoQuick precipitation reagent and analyzed by quantification of the liver-specific RNAs albumin and microRNA-122 (miR-122). In vivo, significant elevations in circulating exosomal albumin mRNA were observed at subtoxic APAP exposures. Significant increases in exosomal albumin mRNA were also observed in primary rat hepatocytes at subtoxic APAP concentrations. In primary human hepatocytes, APAP elicited increases in both exosomal albumin mRNA and exosomal miR-122 without overt cytotoxicity. However, the number of HDE produced in vitro in response to APAP did not increase with exosomal RNA quantity. We conclude that significant drug-induced alterations in the liver-specific RNA content of HDE occur at subtoxic APAP exposures in vivo and in vitro, and that these changes appear to reflect selective packaging rather than changes in exosome number. The current findings demonstrate that translationally relevant HDE alterations occur in the absence of overt hepatocellular toxicity, and support the hypothesis that HDE released by stressed hepatocytes may mediate early immune responses in DILI.
Collapse
Affiliation(s)
- Natalie S Holman
- *Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709
| | - Merrie Mosedale
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709
| | - Kristina K Wolf
- Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709; QPS DMPK Hepatic Biosciences, Research Triangle Park, North Carolina 27709
| | - Edward L LeCluyse
- *Curriculum in Toxicology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709
| | - Paul B Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; Institute for Drug Safety Sciences, University of North Carolina at Chapel Hill, Research Triangle Park, North Carolina 27709;
| |
Collapse
|
271
|
Zhang J, Xu P, Song P, Wang H, Zhang Y, Hu Q, Wang G, Zhang S, Yu Q, Billiar TR, Wang C, Zhang J. CCL2-CCR2 signaling promotes hepatic ischemia/reperfusion injury. J Surg Res 2016; 202:352-62. [PMID: 27229110 DOI: 10.1016/j.jss.2016.02.029] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Revised: 01/30/2016] [Accepted: 02/24/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND Liver ischemia/reperfusion (I/R) injury is a type of uncontrolled inflammatory cascade in which neutrophils, an early infiltrating immune cell population, elicit significant tissue damage. However, the precise mechanism for neutrophil recruitment and infiltration remains to be fully characterized. METHODS A hepatic partial I/R model was reproduced in wild-type, CCL2(-/-) and CCR2(-/-) mice. Tissue damage was evaluated by serum enzyme analysis, hematoxylin-eosin staining, and cytokine production measurement. Mobilization of neutrophils from the bone marrow and subsequent infiltration into the liver were measured by flow cytometry. C-C motif chemokine receptor 2 (CCR2) expression on neutrophils and C-C motif chemokine ligand 2 (CCL2) chemotaxis were measured using flow cytometry. The cellular source of CCL2 in the liver was determined by deleting specific cell groups and performing intracellular staining. RESULTS Liver damage was ameliorated, and neutrophil recruitment and accumulation were decreased in both CCL2(-/-) and CCR2(-/-) mice compared with wild-type mice. Neutrophils displayed upregulated expression of CCR2 during I/R, and these cells were required for CCL2-induced chemotaxis. Depletion of Kupffer cells protected the liver from I/R injury. Furthermore, genetic ablation of CCL2 reduced liver injury, as demonstrated by decreases in the levels of alanine aminotransferase and aspartate aminotransferase and subsequent reductions in neutrophil recruitment and accumulation. CONCLUSIONS Kupffer cells secrete CCL2 to promote CCR2-expressing neutrophil recruitment from the bone marrow and subsequent infiltration into the liver during I/R. These findings reveal a novel pro-inflammatory role of cell-mediated CCL2-CCR2 interactions during this sterile insult.
Collapse
Affiliation(s)
- Junbin Zhang
- Department of Emergency Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Xu
- Department of Emergency Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Song
- Department of Vascular Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Wang
- Department of Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Zhang
- Department of General Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qinggang Hu
- Department of General Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guoliang Wang
- Department of General Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qilin Yu
- The Center for Biomedical Research, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Timothy R Billiar
- Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Congyi Wang
- The Center for Biomedical Research, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| | - Jinxiang Zhang
- Department of Emergency Surgery, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
272
|
Monsel A, Calfee CS. Focusing on the alveolar epithelium: Alveolar fluid clearance in diffuse versus focal acute respiratory distress syndrome. Anaesth Crit Care Pain Med 2016; 35:75-7. [PMID: 26924611 DOI: 10.1016/j.accpm.2016.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Antoine Monsel
- Multidisciplinary intensive care unit, department of anaesthesiology and critical care, UPMC université, La Pitié-Salpêtrière hospital, Assistance publique-Hôpitaux de Paris, Paris, France.
| | - Carolyn S Calfee
- Departments of medicine and anaesthesia, division of pulmonary and critical care medicine, university of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
273
|
Abstract
The liver is a central immunological organ with a high exposure to circulating antigens and endotoxins from the gut microbiota, particularly enriched for innate immune cells (macrophages, innate lymphoid cells, mucosal-associated invariant T (MAIT) cells). In homeostasis, many mechanisms ensure suppression of immune responses, resulting in tolerance. Tolerance is also relevant for chronic persistence of hepatotropic viruses or allograft acceptance after liver transplantation. The liver can rapidly activate immunity in response to infections or tissue damage. Depending on the underlying liver disease, such as viral hepatitis, cholestasis or NASH, different triggers mediate immune-cell activation. Conserved mechanisms such as molecular danger patterns (alarmins), Toll-like receptor signalling or inflammasome activation initiate inflammatory responses in the liver. The inflammatory activation of hepatic stellate and Kupffer cells results in the chemokine-mediated infiltration of neutrophils, monocytes, natural killer (NK) and natural killer T (NKT) cells. The ultimate outcome of the intrahepatic immune response (for example, fibrosis or resolution) depends on the functional diversity of macrophages and dendritic cells, but also on the balance between pro-inflammatory and anti-inflammatory T-cell populations. As reviewed here, tremendous progress has helped to understand the fine-tuning of immune responses in the liver from homeostasis to disease, indicating promising targets for future therapies in acute and chronic liver diseases.
Collapse
Affiliation(s)
- Felix Heymann
- Department of Medicine III, RWTH University-Hospital Aachen, Pauwelsstrasse 30, Aachen 52074, Germany
| | - Frank Tacke
- Department of Medicine III, RWTH University-Hospital Aachen, Pauwelsstrasse 30, Aachen 52074, Germany
| |
Collapse
|
274
|
Yu B, Li X, Wan Q, Han W, Deng C, Guo C. High-Mobility Group Box-1 Protein Disrupts Alveolar Elastogenesis of Hyperoxia-Injured Newborn Lungs. J Interferon Cytokine Res 2016; 36:159-68. [PMID: 26982166 DOI: 10.1089/jir.2015.0080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although high-mobility group box-1 (HMGB1) levels in tracheal aspirates are associated with the pathological features of bronchopulmonary dysplasia (BPD), the role of HMGB1 in the terminal stage of abnormal alveologenesis has not yet been understood. In this study, we addressed the role of HMGB1 in the elastogenesis disruption in the lungs of newborn mice with BPD. We found that elevations of whole lung HMGB1 level were associated with impaired alveolar development and aberrant elastin production in 85% O2-exposed lungs. HMGB1 neutralizing antibody attenuated the structural disintegration developed in hyperoxia-damaged lungs. Furthermore, HMGB1 inhibition rescued the neutrophil influx in hyperoxia-injured lung and partially abolished the mRNA level of the proinflammatory mediators, interleukin (IL)-1β and transforming growth factor (TGF)-β1. These data suggested that pulmonary HMGB1 plays an important role in the disruption of elastogenesis in the terminal stage of lung development through reduced pulmonary inflammatory response.
Collapse
Affiliation(s)
- Benli Yu
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Xiaoyu Li
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Qiufeng Wan
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Wenli Han
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,3 Department of Pharmacology, Chongqing Medical University , Chongqing, P.R. China
| | - Chun Deng
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| | - Chunbao Guo
- 1 Department of Neonatology, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,2 Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China .,4 Department of Hepatology and Liver Transplantation Center, Children's Hospital of Chongqing Medical University , Chongqing, P.R. China
| |
Collapse
|
275
|
Zhang H, Liu Z, Liu S. HMGB1 induced inflammatory effect is blocked by CRISPLD2 via MiR155 in hepatic fibrogenesis. Mol Immunol 2016; 69:1-6. [DOI: 10.1016/j.molimm.2015.10.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Revised: 10/23/2015] [Accepted: 10/26/2015] [Indexed: 12/15/2022]
|
276
|
Stutchfield BM, Antoine DJ, Mackinnon AC, Gow DJ, Bain CC, Hawley CA, Hughes MJ, Francis B, Wojtacha D, Man TY, Dear JW, Devey LR, Mowat AM, Pollard JW, Park BK, Jenkins SJ, Simpson KJ, Hume DA, Wigmore SJ, Forbes SJ. CSF1 Restores Innate Immunity After Liver Injury in Mice and Serum Levels Indicate Outcomes of Patients With Acute Liver Failure. Gastroenterology 2015; 149:1896-1909.e14. [PMID: 26344055 PMCID: PMC4672154 DOI: 10.1053/j.gastro.2015.08.053] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 08/01/2015] [Accepted: 08/27/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Liver regeneration requires functional liver macrophages, which provide an immune barrier that is compromised after liver injury. The numbers of liver macrophages are controlled by macrophage colony-stimulating factor (CSF1). We examined the prognostic significance of the serum level of CSF1 in patients with acute liver injury and studied its effects in mice. METHODS We measured levels of CSF1 in serum samples collected from 55 patients who underwent partial hepatectomy at the Royal Infirmary Edinburgh between December 2012 and October 2013, as well as from 78 patients with acetaminophen-induced acute liver failure admitted to the Royal Infirmary Edinburgh or the University of Kansas Medical Centre. We studied the effects of increased levels of CSF1 in uninjured mice that express wild-type CSF1 receptor or a constitutive or inducible CSF1-receptor reporter, as well as in chemokine receptor 2 (Ccr2)-/- mice; we performed fate-tracing experiments using bone marrow chimeras. We administered CSF1-Fc (fragment, crystallizable) to mice after partial hepatectomy and acetaminophen intoxication, and measured regenerative parameters and innate immunity by clearance of fluorescent microbeads and bacterial particles. RESULTS Serum levels of CSF1 increased in patients undergoing liver surgery in proportion to the extent of liver resected. In patients with acetaminophen-induced acute liver failure, a low serum level of CSF1 was associated with increased mortality. In mice, administration of CSF1-Fc promoted hepatic macrophage accumulation via proliferation of resident macrophages and recruitment of monocytes. CSF1-Fc also promoted transdifferentiation of infiltrating monocytes into cells with a hepatic macrophage phenotype. CSF1-Fc increased innate immunity in mice after partial hepatectomy or acetaminophen-induced injury, with resident hepatic macrophage as the main effector cells. CONCLUSIONS Serum CSF1 appears to be a prognostic marker for patients with acute liver injury. CSF1 might be developed as a therapeutic agent to restore innate immune function after liver injury.
Collapse
Affiliation(s)
- Benjamin M. Stutchfield
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom,Division of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Daniel J. Antoine
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alison C. Mackinnon
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Deborah J. Gow
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Calum C. Bain
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Catherine A. Hawley
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael J. Hughes
- Division of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Benjamin Francis
- Department of Biostatistics, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Davina Wojtacha
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Tak Y. Man
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - James W. Dear
- National Poisons Information Service Edinburgh, Royal Infirmary of Edinburgh, University of Edinburgh, Edinburgh, United Kingdom
| | - Luke R. Devey
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Alan M. Mowat
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Jeffrey W. Pollard
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - B. Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Jenkins
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Kenneth J. Simpson
- Division of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Hume
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J. Wigmore
- Division of Clinical and Surgical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Stuart J. Forbes
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, United Kingdom,Reprint requests Address requests for reprints to: S. J. Forbes, MD, Scottish Centre for Regenerative Medicine, 5 Little France Drive, Edinburgh BioQuarter, Edinburgh EH16 4UU, United Kingdom. fax: (44) (0)131-651-9501.
| |
Collapse
|
277
|
Osumi W, Jin D, Imai Y, Tashiro K, Li ZL, Otsuki Y, Maemura K, Komeda K, Hirokawa F, Hayashi M, Takai S, Uchiyama K. Recombinant human soluble thrombomodulin improved lipopolysaccharide/d-galactosamine-induced acute liver failure in mice. J Pharmacol Sci 2015; 129:233-9. [DOI: 10.1016/j.jphs.2015.11.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/30/2015] [Accepted: 11/18/2015] [Indexed: 01/27/2023] Open
|
278
|
Role of inflammatory cells in fibroblast activation. J Mol Cell Cardiol 2015; 93:143-8. [PMID: 26593723 DOI: 10.1016/j.yjmcc.2015.11.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/12/2015] [Accepted: 11/14/2015] [Indexed: 11/21/2022]
Abstract
Although fibrosis is an essential response to acute cardiac tissue injury, prolonged myofibroblast activation and progressive fibrosis lead to further distortion of tissue architecture and worsened cardiac function. Thus, optimal tissue repair following injury requires tight control over myofibroblast activation. It is now recognized that inflammation plays a critical role in regulating fibrosis. In this review we will highlight how advances in the field of innate immunity have led to a better understanding of the role of inflammation in cardiovascular disease and, in particular, in the regulation of fibrosis. Specifically, we will discuss how the innate immune system recognizes tissue damage in settings of acute injury and chronic cardiovascular disease. We will also review the role of different cell populations in this response, particularly the unique role of different macrophage subsets and mast cells.
Collapse
|
279
|
Wang C, de Souza AW, Westra J, Bijl M, Chen M, Zhao MH, Kallenberg CG. Emerging role of high mobility group box 1 in ANCA-associated vasculitis. Autoimmun Rev 2015. [DOI: 10.1016/j.autrev.2015.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
280
|
Cell Death Conversion under Hypoxic Condition in Tumor Development and Therapy. Int J Mol Sci 2015; 16:25536-51. [PMID: 26512660 PMCID: PMC4632814 DOI: 10.3390/ijms161025536] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 10/07/2015] [Accepted: 10/19/2015] [Indexed: 12/29/2022] Open
Abstract
Hypoxia, which is common during tumor progression, plays important roles in tumor biology. Failure in cell death in response to hypoxia contributes to progression and metastasis of tumors. On the one hand, the metabolic and oxidative stress following hypoxia could lead to cell death by triggering signal cascades, like LKB1/AMPK, PI3K/AKT/mTOR, and altering the levels of effective components, such as the Bcl-2 family, Atg and p62. On the other hand, hypoxia-induced autophagy can serve as a mechanism to turn over nutrients, so as to mitigate the adverse condition and then avoid cell death potentially. Due to the effective role of hypoxia, this review focuses on the crosstalk in cell death under hypoxia in tumor progression. Additionally, the illumination of cell death in hypoxia could shed light on the clinical applications of cell death targeted therapy.
Collapse
|
281
|
Matthay MA, Ware LB. Resolution of Alveolar Edema in Acute Respiratory Distress Syndrome. Physiology and Biology. Am J Respir Crit Care Med 2015; 192:124-5. [PMID: 26177166 DOI: 10.1164/rccm.201505-0938ed] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Michael A Matthay
- 1 Cardiovascular Research Institute University of San Francisco San Francisco, California
| | - Lorraine B Ware
- 2 Department of Medicine Vanderbilt University School of Medicine Nashville, Tennessee
| |
Collapse
|
282
|
Mitochondrial ROS Induces Cardiac Inflammation via a Pathway through mtDNA Damage in a Pneumonia-Related Sepsis Model. PLoS One 2015; 10:e0139416. [PMID: 26448624 PMCID: PMC4598156 DOI: 10.1371/journal.pone.0139416] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022] Open
Abstract
We have previously shown that mitochondria-targeted vitamin E (Mito-Vit-E), a mtROS specific antioxidant, improves cardiac performance and attenuates inflammation in a pneumonia-related sepsis model. In this study, we applied the same approaches to decipher the signaling pathway(s) of mtROS-dependent cardiac inflammation after sepsis. Sepsis was induced in Sprague Dawley rats by intratracheal injection of S. pneumoniae. Mito-Vit-E, vitamin E or vehicle was administered 30 minutes later. In myocardium 24 hours post-inoculation, Mito-Vit-E, but not vitamin E, significantly protected mtDNA integrity and decreased mtDNA damage. Mito-Vit-E alleviated sepsis-induced reduction in mitochondria-localized DNA repair enzymes including DNA polymerase γ, AP endonuclease, 8-oxoguanine glycosylase, and uracil-DNA glycosylase. Mito-Vit-E dramatically improved metabolism and membrane integrity in mitochondria, suppressed leakage of mtDNA into the cytoplasm, inhibited up-regulation of Toll-like receptor 9 (TLR9) pathway factors MYD88 and RAGE, and limited RAGE interaction with its ligand TFAM in septic hearts. Mito-Vit-E also deactivated NF-κB and caspase 1, reduced expression of the essential inflammasome component ASC, and decreased inflammatory cytokine IL–1β. In vitro, both Mito-Vit-E and TLR9 inhibitor OND-I suppressed LPS-induced up-regulation in MYD88, RAGE, ASC, active caspase 1, and IL–1β in cardiomyocytes. Since free mtDNA escaped from damaged mitochondria function as a type of DAMPs to stimulate inflammation through TLR9, these data together suggest that sepsis-induced cardiac inflammation is mediated, at least partially, through mtDNA-TLR9-RAGE. At last, Mito-Vit-E reduced the circulation of myocardial injury marker troponin-I, diminished apoptosis and amended morphology in septic hearts, suggesting that mitochondria-targeted antioxidants are a potential cardioprotective approach for sepsis.
Collapse
|
283
|
TGF-β in Hepatic Stellate Cell Activation and Liver Fibrogenesis: Updated. CURRENT PATHOBIOLOGY REPORTS 2015. [DOI: 10.1007/s40139-015-0089-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
284
|
Abstract
High mobility group box 1 (HMGB1) plays a key role in human health and disease. Currently, three different labs in the USA use Cre-lox technology to create mice with a hepatocyte-specific Hmgb1 deletion (HMGB1-HC-KO mice) by backcrossing Hmgb1 (flox)/(flox) mice to Alb-Cre mice. This mouse strain has a different phenotype following exposure to several stressors.
Collapse
Affiliation(s)
- Xiaofang Sun
- a The Third Affiliated Hospital of Guangzhou Medical University ; Guangzhou , Guangdong , China
| | | |
Collapse
|
285
|
Abstract
HMGB1 (high mobility group box 1), a ubiquitously expressed DNA-binding nucleoprotein, has not only been attributed with important functions in the regulation of gene expression but is thought to function as an important damage-associated molecular pattern in the extracellular space. Recently, conditional Hmgb1 deletion strategies have been employed to overcome the perinatal mortality of global Hmgb1 deletion and to understand HMGB1 functions under disease conditions. From these studies, it has become evident that HMGB1 is not required for normal organ function. However, the different conditional ablation strategies have yielded contradictory results in some disease models. With nearly complete recombination in all transgenic mouse models, the main reason for opposite results is likely to lie within different targeting strategies. In summary, different targeting strategies need to be taken into account when interpreting HMGB1 functions, and further efforts need to be undertaken to compare these models side by side.
Collapse
Affiliation(s)
- Peter Huebener
- a I. Medizinische Klinik und Poliklinik; Universitätsklinikum Hamburg-Eppendorf ; Hamburg , Germany
| | | | | |
Collapse
|
286
|
Wang X, Xiang L, Li H, Chen P, Feng Y, Zhang J, Yang N, Li F, Wang Y, Zhang Q, Li F, Cao F. The Role of HMGB1 Signaling Pathway in the Development and Progression of Hepatocellular Carcinoma: A Review. Int J Mol Sci 2015; 16:22527-40. [PMID: 26393575 PMCID: PMC4613322 DOI: 10.3390/ijms160922527] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 12/15/2022] Open
Abstract
The story of high mobility group protein B1 (HMGB1) in cancer is complicated and the function of HMGB1 in different cancers is uncertain. This review aims to retrieve literature regarding HMGB1 from English electronic resources, analyze and summarize the role of the HMGB1 signaling pathway in hepatocellular carcinoma (HCC), and provide useful information for carcinogenesis and progression of HCC. Results showed that HMGB1 could induce cell proliferation, differentiation, cell death, angiogenesis, metastasis, inflammation, and enhance immunofunction in in vitro and in vivo HCC models. HMGB1 and its downstream receptors RAGE, TLRs and TREM-1 may be potential anticancer targets. In conclusion, HMGB1 plays an important role in oncogenesis and represents a novel therapeutic target, which deserves further study.
Collapse
Affiliation(s)
- Xuanbin Wang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Longchao Xiang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Hongliang Li
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Ping Chen
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Yibin Feng
- School of Chinese Medicine, the University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China.
| | - Jingxuan Zhang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Nian Yang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Fei Li
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Ye Wang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Quifang Zhang
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
- Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Fang Li
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| | - Fengjun Cao
- Laboratory of Chinese Herbal Pharmacology, Renmin Hospital, 30 South Renmin Road, Shiyan 442000, Hubei, China.
| |
Collapse
|
287
|
Geng Y, Ma Q, Liu YN, Peng N, Yuan FF, Li XG, Li M, Wu YS, Li BL, Song WB, Zhu W, Xu WW, Fan J, Su L. Heatstroke induces liver injury via IL-1β and HMGB1-induced pyroptosis. J Hepatol 2015; 63:622-33. [PMID: 25931416 DOI: 10.1016/j.jhep.2015.04.010] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 03/28/2015] [Accepted: 04/02/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Liver injury is a common complication of heat stroke (HS), and often constitutes a direct cause for patient death. The cellular and molecular mechanism underlying HS-induced liver injury remains unclear. Recent evidence indicates that inflammasome plays an important role in mediating sterile inflammation triggered by tissue damage. Using a rat HS model, we identified a novel mechanism by which inflammasome-dependent interleukin-1β (IL-1β) activation and hepatocyte pyroptosis mediate HS-induced liver injury. METHODS To induce HS, rats were subjected to heat exposure. Inhibition of inflammasomes was achieved by RNA silencing and pharmacologic inhibitor prior to heat exposure. Inflammasome assembly, caspase-1 activation, histological changes, as well as serum levels of liver enzymes were measured. RESULTS We demonstrated that the onset of HS activated inflammasome in the liver as evidenced by increased capase-1 activity and the association of inflammasome components NOD-like receptor family pyrin domain containing 3 (Nlrp3) and apoptosis speck-like protein containing a caspase-recruitment domain (ASC); and the activated inflammasome, in turn, induced IL-1β activation and hepatocyte pyroptosis, and subsequent augmented liver injury. HS-induced hepatocyte inflammasome activation seems to be high-mobility group box 1 (HMGB1) dependent. Inhibition of Nlrp3, caspase-1, or HMGB1 prevented HS-induced liver inflammation and ameliorated liver injury. CONCLUSIONS These findings demonstrate an important role of HMGB1 in mediating inflammasome activation in the development of liver injury following HS, and suggest that targeting inflammasome may represent a novel therapeutic strategy to limit cell death and prevent liver failure after HS.
Collapse
Affiliation(s)
- Yan Geng
- Department of Gastroenterology, 303 Hospital of People's Liberation Army, Nanning 530021, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China.
| | - Qiang Ma
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China.
| | - Ya-Nan Liu
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China
| | - Na Peng
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Fang-Fang Yuan
- Department of Graduate School, Southern Medical University, Guangzhou 510515, PR China; Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xing-Gui Li
- Department of Neurology, The Fifth People's Hospital of Chongqing, Chongqing 400062, PR China; Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Ming Li
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Ying-Song Wu
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Bing-ling Li
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Wei-bing Song
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China
| | - Wei Zhu
- Department of Toxicology, Guangzhou Center for Disease Control and Prevention, Guangzhou 510515, PR China
| | - Wei-Wen Xu
- State Key Laboratory of Organ Failure Research, Institute of Antibody Engineering, Southern Medical University, Guangzhou 510515, PR China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | - Lei Su
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou 510010, PR China.
| |
Collapse
|
288
|
Vliegenthart ADB, Antoine DJ, Dear JW. Target biomarker profile for the clinical management of paracetamol overdose. Br J Clin Pharmacol 2015; 80:351-62. [PMID: 26076366 DOI: 10.1111/bcp.12699] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/03/2015] [Accepted: 06/09/2015] [Indexed: 12/21/2022] Open
Abstract
Paracetamol (acetaminophen) overdose is one of the most common causes of acute liver injury in the Western world. To improve patient care and reduce pressure on already stretched health care providers new biomarkers are needed that identify or exclude liver injury soon after an overdose of paracetamol is ingested. This review highlights the current state of paracetamol poisoning management and how novel biomarkers could improve patient care and save healthcare providers money. Based on the widely used concept of defining a target product profile, a target biomarker profile is proposed that identifies desirable and acceptable key properties for a biomarker in development to enable the improved treatment of this patient population. The current biomarker candidates, with improved hepatic specificity and based on the fundamental mechanistic basis of paracetamol-induced liver injury, are reviewed and their performance compared with our target profile.
Collapse
Affiliation(s)
- A D Bastiaan Vliegenthart
- Pharmacology, Toxicology & Therapeutics, University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh
| | - Daniel J Antoine
- MRC Centre for Drug Safety Science, Department of Molecular & Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - James W Dear
- Pharmacology, Toxicology & Therapeutics, University/BHF Centre for Cardiovascular Science, University of Edinburgh, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh
| |
Collapse
|
289
|
Liu Z, Du R, Long J, Guo K, Ge C, Bi S, Xu Y. microRNA-218 promotes gemcitabine sensitivity in human pancreatic cancer cells by regulating HMGB1 expression. Chin J Cancer Res 2015; 27:267-78. [PMID: 26157323 DOI: 10.3978/j.issn.1000-9604.2015.04.06] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/24/2015] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE The purpose of this study was to examine the effect of gemcitabine (GEM) on microRNA-218 (miR-218) expression in human pancreatic cancer cells. METHODS Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to examine the differences in miR-218 expression between the GEM-sensitive BxPC-3 pancreatic cancer cells and GEM-resistant PANC-1 cells. The effect of GEM on the expression of miR-218 in PANC-1 cells was also investigated. PANC-1 cells were transfected either with HMGB1 siRNA to knock down the expression of HMGB1 or with the recombinant HMGB1 expression vector (pcDNA3.1-HMGB1) to overexpress HMGB1. The effect of ectopic expression of HMGB1 on the apoptosis of miR-218-transfected and GEM-treated PANC-1 cells was examined by flow cytometric analysis. RESULTS The miR-218 expression level was lower in GEM-resistant PANC-1 cells compared to GEM-sensitive BxPC-3 cells (P<0.05). The percentage of apoptotic PANC-1 cells was significantly increased in the miR-218 mimic + GEM group compared to the mimic ctrl + GEM group and the normal control group (P<0.01). The HMGB1 expression level was markedly decreased in PANC-1 cells transfected with HMGB1 siRNA but was significantly increased in PANC-1 cells transfected with the recombinant HMGB1 expression vector, pcDNA3.1-HMGB1 (P<0.01). The proportion of apoptotic PANC-1 cells was significantly lower in the miR-218 mimic + GEM + pcDNA3.1-HMGB1 group compared to the miR-218 mimic + GEM + HMGB1 siRNA group (P<0.01). CONCLUSIONS The expression level of miR-218 was downregulated in the GEM-resistant cell line. miR-218 promoted the sensitivity of PANC-1 cells to GEM, which was achieved mainly through regulating the expression of HMGB1 in PANC-1 cells.
Collapse
Affiliation(s)
- Zhe Liu
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Ruixia Du
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Jin Long
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Kejian Guo
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Chunlin Ge
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Shulong Bi
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| | - Yuanhong Xu
- 1 Department of Pancreatic Surgery, First Hospital of China Medical University, Shenyang 110001, China ; 2 Department of Otorhinolaryngology, Fengtian Hospital, Shenyang Medical University, Shenyang 110024, China
| |
Collapse
|
290
|
Zhang Z, Wang M, Zhou L, Feng X, Cheng J, Yu Y, Gong Y, Zhu Y, Li C, Tian L, Huang Q. Increased HMGB1 and cleaved caspase-3 stimulate the proliferation of tumor cells and are correlated with the poor prognosis in colorectal cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:51. [PMID: 25986235 PMCID: PMC4446854 DOI: 10.1186/s13046-015-0166-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 05/06/2015] [Indexed: 12/21/2022]
Abstract
Background Dying tumor cells after irradiation could promote the proliferation of living tumor cells might cause tumor relapse and treatment failure. Our previous study showed that activated caspase-3 after irradiation probably participates in tumor repopulation. In this study, we investigated whether high mobility group box 1(HMGB1) is also involved in tumor repopulation. Methods Colorectal tumor cells were irradiated. The cleaved caspase-3 (CC3) in irradiated tumor cells and HMGB1 in the supernatant of irradiated tumor cells were detected by Western blot. A large number of irradiated colorectal tumor cells (feeder cells) were then co-cultured with a small number of luciferase-labeled living colorectal tumor cells (reporter cells) and proliferation of reporter cells was measured by bioluminescence imaging. The CC3 and HMGB1 protein expression in colorectal tumor and peritumoral tissues were detected by immunohistochemistry and their correlation with prognosis were analyzed. Results The irradiated colorectal tumor cells underwent apoptosis and necrosis and produced CC3 in tumor cells and HMGB1 in the supernatant of cultured cells. The increased expression of secretory HMGB1 correlated with CC3 level and proliferating cell nuclear antigen (PCNA) after irradiation in vitro. The irradiated dying cells remarkably stimulated living tumor cell proliferation. Interestedly, immunohistochemistry staining showed that positive HMGB1, CC3, and Ki67 expression were significantly higher in colorectal tumor tissues than in peritumoral tissues (p <0.01). The Kaplan-Meier survival analysis revealed that high HMGB1, CC3, and Ki67 levels were significantly associated with poor prognosis (p <0.05, p <0.01). Multivariate analysis using Cox proportional hazards model showed that TNM staging and HMGB1 were independent prognostic factors in patients with colorectal cancer (CRC) (p <0.01, p <0.001). Conclusion Both apoptotic and necrotic cells could stimulate proliferation of living tumor cells, and the increased expression of CC3 and HMGB1 in tumor cells could be new markers for poor prognosis in colorectal cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0166-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhengxiang Zhang
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Min Wang
- The Department of Surgery, The Branch Hospital of Shanghai General Hospital, Shanghai, 200080, China.
| | - Ling Zhou
- The Department of Surgery, The Branch Hospital of Shanghai General Hospital, Shanghai, 200080, China.
| | - Xiao Feng
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Jin Cheng
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Yang Yu
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Yanping Gong
- Experimental Research Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Ying Zhu
- The Department of Surgery, The Branch Hospital of Shanghai General Hospital, Shanghai, 200080, China.
| | - Chuanyuan Li
- Department of Dermatology, Medical Center, Duke University, Durham, NC, 27710, USA.
| | - Ling Tian
- Experimental Research Center, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| | - Qian Huang
- The Comprehensive Cancer Center & Shanghai Key Laboratory for Pancreatic Diseases, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 201620, China.
| |
Collapse
|
291
|
Maile R, Jones S, Pan Y, Zhou H, Jaspers I, Peden DB, Cairns BA, Noah TL. Association between early airway damage-associated molecular patterns and subsequent bacterial infection in patients with inhalational and burn injury. Am J Physiol Lung Cell Mol Physiol 2015; 308:L855-60. [PMID: 25770180 DOI: 10.1152/ajplung.00321.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/10/2015] [Indexed: 01/24/2023] Open
Abstract
Bacterial infection is a major cause of morbidity affecting outcome following burn and inhalation injury. While experimental burn and inhalation injury animal models have suggested that mediators of cell damage and inflammation increase the risk of infection, few studies have been done on humans. This is a prospective, observational study of patients admitted to the North Carolina Jaycee Burn Center at the University of North Carolina who were intubated and on mechanical ventilation for treatment of burn and inhalational injury. Subjects were enrolled over a 2-yr period and followed till discharge or death. Serial bronchial washings from clinically indicated bronchoscopies were collected and analyzed for markers of tissue injury and inflammation. These include damage-associated molecular patterns (DAMPs) such as hyaluronic acid (HA), double-stranded DNA (dsDNA), heat-shock protein 70 (HSP-70), and high-mobility group protein B-1 (HMGB-1). The study population was comprised of 72 patients who had bacterial cultures obtained for clinical indications. Elevated HA, dsDNA, and IL-10 levels in bronchial washings obtained early (the first 72 h after injury) were significantly associated with positive bacterial respiratory cultures obtained during the first 14 days postinjury. Independent of initial inhalation injury severity and extent of surface burn, elevated levels of HA dsDNA and IL-10 in the central airways obtained early after injury are associated with subsequent positive bacterial respiratory cultures in patients intubated after acute burn/inhalation injury.
Collapse
Affiliation(s)
- Robert Maile
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina;
| | - Samuel Jones
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina
| | - Yinghao Pan
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Haibo Zhou
- Department of Biostatistics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ilona Jaspers
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - David B Peden
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| | - Bruce A Cairns
- Department of Surgery, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; North Carolina Jaycee Burn Center, Chapel Hill, North Carolina
| | - Terry L Noah
- Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Center for Environmental Medicine, Asthma and Lung Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; and
| |
Collapse
|