1
|
Sen K, Kumar Das S, Ghosh N, Sinha K, Sil PC. Lupeol: A dietary and medicinal triterpene with therapeutic potential. Biochem Pharmacol 2024; 229:116545. [PMID: 39293501 DOI: 10.1016/j.bcp.2024.116545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Lupeol, a triterpene derived from various plants, has emerged as a potent dietary supplement with extensive therapeutic potential. This review offers a comprehensive examination of lupeol's applications across diverse health conditions. By meticulously analyzing current scientific literature, we have synthesized findings that underscore lupeol's impact on cancer, diabetes, gastrointestinal disorders, neurological diseases, dermatological conditions, nephrological issues, and cardiovascular health. The review delves into molecular studies that reveal lupeol's ability to modulate disease pathways and alleviate symptoms, positioning it as a promising therapeutic agent. Moreover, we discuss the potential role of lupeol in clinical practice and public health strategies, emphasizing its substantial benefits as a natural compound. This thorough analysis serves as a critical resource for researchers, providing insights into the multifaceted therapeutic properties of lupeol and its potential to significantly enhance health outcomes.
Collapse
Affiliation(s)
- Koushik Sen
- Jhargram Raj College, Jhargram 721507, India
| | | | | | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India.
| |
Collapse
|
2
|
Grayck MR, McCarthy WC, Solar M, Balasubramaniyan N, Zheng L, Orlicky DJ, Wright CJ. Implications of neonatal absence of innate immune mediated NFκB/AP1 signaling in the murine liver. Pediatr Res 2024; 95:1791-1802. [PMID: 38396130 DOI: 10.1038/s41390-024-03071-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/03/2024] [Accepted: 01/20/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND The developmental immaturity of the innate immune system helps explains the increased risk of infection in the neonatal period. Importantly, innate immune signaling pathways such as p65/NFκB and c-Jun/AP1 are responsible for the prevention of hepatocyte apoptosis in adult animals, yet whether developmental immaturity of these pathways increases the risk of hepatic injury in the neonatal period is unknown. METHODS Using a murine model of endotoxemia (LPS 5 mg/kg IP x 1) in neonatal (P3) and adult mice, we evaluated histologic evidence of hepatic injury and apoptosis, presence of p65/NFκB and c-Jun/AP1 activation and associated transcriptional regulation of apoptotic genes. RESULTS We demonstrate that in contrast to adults, endotoxemic neonatal (P3) mice exhibit a significant increase in hepatic apoptosis. This is associated with absent hepatic p65/NFκB signaling and impaired expression of anti-apoptotic target genes. Hepatic c-Jun/AP1 activity was attenuated in endotoxemic P3 mice, with resulting upregulation of pro-apoptotic factors. CONCLUSIONS These results demonstrate that developmental absence of innate immune p65/NFκB and c-Jun/AP1 signaling, and target gene expression is associated with apoptotic injury in neonatal mice. More work is needed to determine if this contributes to long-term hepatic dysfunction, and whether immunomodulatory approaches can prevent this injury. IMPACT Various aspects of developmental immaturity of the innate immune system may help explain the increased risk of infection in the neonatal period. In adult models of inflammation and infection, innate immune signaling pathways such as p65/NFκB and c-Jun/AP1 are responsible for a protective, pro-inflammatory transcriptome and regulation of apoptosis. We demonstrate that in contrast to adults, endotoxemic neonatal (P3) mice exhibit a significant increase in hepatic apoptosis associated with absent hepatic p65/NFκB signaling and c-Jun/AP1 activity. We believe that these results may explain in part hepatic dysfunction with neonatal sepsis, and that there may be unrecognized developmental and long-term hepatic implications of early life exposure to systemic inflammatory stress.
Collapse
Affiliation(s)
- Maya R Grayck
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - William C McCarthy
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mack Solar
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Natarajan Balasubramaniyan
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Lijun Zheng
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - David J Orlicky
- Dept of Pathology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Clyde J Wright
- Section of Neonatology, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
3
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
4
|
Shapiro L, Scherger S, Franco-Paredes C, Gharamti AA, Fraulino D, Henao-Martinez AF. Chasing the Ghost: Hyperinflammation Does Not Cause Sepsis. Front Pharmacol 2022; 13:910516. [PMID: 35814227 PMCID: PMC9260244 DOI: 10.3389/fphar.2022.910516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
Sepsis is infection sufficient to cause illness in the infected host, and more severe forms of sepsis can result in organ malfunction or death. Severe forms of Coronavirus disease-2019 (COVID-19), or disease following infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are examples of sepsis. Following infection, sepsis is thought to result from excessive inflammation generated in the infected host, also referred to as a cytokine storm. Sepsis can result in organ malfunction or death. Since COVID-19 is an example of sepsis, the hyperinflammation concept has influenced scientific investigation and treatment approaches to COVID-19. However, decades of laboratory study and more than 100 clinical trials designed to quell inflammation have failed to reduce sepsis mortality. We examine theoretical support underlying widespread belief that hyperinflammation or cytokine storm causes sepsis. Our analysis shows substantial weakness of the hyperinflammation approach to sepsis that includes conceptual confusion and failure to establish a cause-and-effect relationship between hyperinflammation and sepsis. We conclude that anti-inflammation approaches to sepsis therapy have little chance of future success. Therefore, anti-inflammation approaches to treat COVID-19 are likewise at high risk for failure. We find persistence of the cytokine storm concept in sepsis perplexing. Although treatment approaches based on the hyperinflammation concept of pathogenesis have failed, the concept has shown remarkable resilience and appears to be unfalsifiable. An approach to understanding this resilience is to consider the hyperinflammation or cytokine storm concept an example of a scientific paradigm. Thomas Kuhn developed the idea that paradigms generate rules of investigation that both shape and restrict scientific progress. Intrinsic features of scientific paradigms include resistance to falsification in the face of contradictory data and inability of experimentation to generate alternatives to a failing paradigm. We call for rejection of the concept that hyperinflammation or cytokine storm causes sepsis. Using the hyperinflammation or cytokine storm paradigm to guide COVID-19 treatments is likewise unlikely to provide progress. Resources should be redirected to more promising avenues of investigation and treatment.
Collapse
Affiliation(s)
- Leland Shapiro
- Division of Infectious Diseases, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sias Scherger
- Division of Infectious Diseases, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, United States
| | - Carlos Franco-Paredes
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Hospital Infantil de México, Federico Gomez, Mexico City, Mexico
| | - Amal A. Gharamti
- Department of Internal Medicine, Yale University, Waterbury, CT, United States
| | - David Fraulino
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Andrés F. Henao-Martinez
- Division of Infectious Diseases, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
5
|
Tiegs G, Horst AK. TNF in the liver: targeting a central player in inflammation. Semin Immunopathol 2022; 44:445-459. [PMID: 35122118 PMCID: PMC9256556 DOI: 10.1007/s00281-022-00910-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/12/2022] [Indexed: 12/11/2022]
Abstract
Tumour necrosis factor-α (TNF) is a multifunctional cytokine. First recognized as an endogenous soluble factor that induces necrosis of solid tumours, TNF became increasingly important as pro-inflammatory cytokine being involved in the immunopathogenesis of several autoimmune diseases. In the liver, TNF induces numerous biological responses such as hepatocyte apoptosis and necroptosis, liver inflammation and regeneration, and autoimmunity, but also progression to hepatocellular carcinoma. Considering these multiple functions of TNF in the liver, we propose anti-TNF therapies that specifically target TNF signalling at the level of its specific receptors.
Collapse
Affiliation(s)
- Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Andrea K Horst
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
6
|
Li F, Yan R, Wu J, Han Z, Qin M, Liu C, Lu Y. An Antioxidant Enzyme Therapeutic for Sepsis. Front Bioeng Biotechnol 2021; 9:800684. [PMID: 34888304 PMCID: PMC8650590 DOI: 10.3389/fbioe.2021.800684] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 11/08/2021] [Indexed: 11/22/2022] Open
Abstract
Sepsis is a systemic inflammatory response syndrome caused by infections that may lead to organ dysfunction with high mortality. With the rapid increase in the aging population and antimicrobial resistance, developing therapeutics for the treatment of sepsis has been an unmet medical need. Excessive production of reactive oxygen species (ROS) during inflammation is associated with the occurrence of sepsis. We report herein a treatment for sepsis based on PEGylated catalase, which can effectively break down hydrogen peroxide, a key component of ROS that is chemically stable and able to diffuse around the tissues and form downstream ROS. PEGylated catalase can effectively regulate the cytokine production by activated leukocytes, suppress the elevated level of AST, ALT, TNF-α, and IL-6 in mice with induced sepsis, and significantly improve the survival rate.
Collapse
Affiliation(s)
- Feifei Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ran Yan
- Department of Chemical and Biomolecular Engineering, The University of California, Los Angeles, CA, United States
| | - Jun Wu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zeren Han
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Meng Qin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Chaoyong Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.,College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yunfeng Lu
- Department of Chemical and Biomolecular Engineering, The University of California, Los Angeles, CA, United States
| |
Collapse
|
7
|
Mochizuki S, Miyamoto N, Sakurai K. Oligonucleotide delivery to antigen presenting cells by using schizophyllan. Drug Metab Pharmacokinet 2021; 42:100434. [PMID: 34896749 DOI: 10.1016/j.dmpk.2021.100434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/15/2022]
Abstract
Schizophyllan (SPG), a member of the β-glucan family, can form novel complexes with homo-polynucleotides such as poly(dA) through hydrogen bonding between two main chain glucoses and the one nucleotide base. Dectin-1, one of the major receptors for β-glucans, is known to be expressed on antigen presenting cells (APCs) such as macrophages and dendritic cells. This suggests that the above-mentioned complexes could deliver bound functional oligonucleotides (ODNs) including antisense (AS)-ODNs, small interfering RNA, and CpG-ODNs to the APCs. Analysis using a quartz crystal microbalance revealed that a complex consisting of SPG and dA60 with a phosphorothioate backbone was recognized by recombinant Dectin-1 protein. Treatment with this complex containing an AS-ODN for tumor necrosis factor alpha protected mice against lipopolysaccharide-induced hepatitis at a very low AS-ODN dose. Moreover, immunization with CpG-ODN/SPG complex and antigenic proteins induced potent antigen specific immune responses. The present review also represents peptide delivery by conjugation with dA60 and the preparation of a nanogel using DNA-DNA hybridization. These findings indicate that the delivery of a specific ODN using β-glucans could be used for treating various diseases caused by APCs and for activating antigen specific immune responses.
Collapse
Affiliation(s)
- Shinichi Mochizuki
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan.
| | - Noriko Miyamoto
- Department of Applied Chemistry, Aichi Institute of Technology, 1247, Yachigusa, Yakusacho, Toyota, Aichi, 470-0392, Japan
| | - Kazuo Sakurai
- Department of Chemistry and Biochemistry, The University of Kitakyushu, 1-1, Hibikino, Wakamatsu-ku, Kitakyushu, Fukuoka, 808-0135, Japan
| |
Collapse
|
8
|
Koschel J, Nishanth G, Just S, Harit K, Kröger A, Deckert M, Naumann M, Schlüter D. OTUB1 prevents lethal hepatocyte necroptosis through stabilization of c-IAP1 during murine liver inflammation. Cell Death Differ 2021; 28:2257-2275. [PMID: 33712742 PMCID: PMC8257688 DOI: 10.1038/s41418-021-00752-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/25/2021] [Accepted: 02/04/2021] [Indexed: 01/31/2023] Open
Abstract
In bacterial and sterile inflammation of the liver, hepatocyte apoptosis is, in contrast to necroptosis, a common feature. The molecular mechanisms preventing hepatocyte necroptosis and the potential consequences of hepatocyte necroptosis are largely unknown. Apoptosis and necroptosis are critically regulated by the ubiquitination of signaling molecules but especially the regulatory function of deubiquitinating enzymes (DUBs) is imperfectly defined. Here, we addressed the role of the DUB OTU domain aldehyde binding-1 (OTUB1) in hepatocyte cell death upon both infection with the hepatocyte-infecting bacterium Listeria monocytogenes (Lm) and D-Galactosamine (DGal)/Tumor necrosis factor (TNF)-induced sterile inflammation. Combined in vivo and in vitro experiments comprising mice lacking OTUB1 specifically in liver parenchymal cells (OTUB1LPC-KO) and human OTUB1-deficient HepG2 cells revealed that OTUB1 prevented hepatocyte necroptosis but not apoptosis upon infection with Lm and DGal/TNF challenge. Lm-induced necroptosis in OTUB1LPC-KO mice resulted in increased alanine aminotransferase (ALT) and lactate dehydrogenase (LDH) release and rapid lethality. Treatment with the receptor-interacting serine/threonine-protein kinase (RIPK) 1 inhibitor necrostatin-1s and deletion of the pseudokinase mixed lineage kinase domain-like protein (MLKL) prevented liver damage and death of infected OTUB1LPC-KO mice. Mechanistically, OTUB1 reduced K48-linked polyubiquitination of the cellular inhibitor of apoptosis 1 (c-IAP1), thereby diminishing its degradation. In the absence of OTUB1, c-IAP1 degradation resulted in reduced K63-linked polyubiquitination and increased phosphorylation of RIPK1, RIPK1/RIPK3 necrosome formation, MLKL-phosphorylation and hepatocyte death. Additionally, OTUB1-deficiency induced RIPK1-dependent extracellular-signal-regulated kinase (ERK) activation and TNF production in Lm-infected hepatocytes. Collectively, these findings identify OTUB1 as a novel regulator of hepatocyte-intrinsic necroptosis and a critical factor for survival of bacterial hepatitis and TNF challenge.
Collapse
Affiliation(s)
- Josephin Koschel
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Gopala Nishanth
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.
- Institute of Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Sissy Just
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Kunjan Harit
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, Magdeburg, Germany
- Innate Immunity and Infection Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Martina Deckert
- Department of Neuropathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.
- Institute of Medical Microbiology and Hospital Hygiene, Otto von Guericke University Magdeburg, Magdeburg, Germany.
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany.
| |
Collapse
|
9
|
Abstract
Complement protein C3dg, a key linkage between innate and adaptive immunity, is capable of stimulating both humoral and cell-mediated immune responses, leading to considerable interest in its use as a molecular adjuvant. However, the potential of C3dg as an adjuvant is limited without ways of controllably assembling multiple copies of it into vaccine platforms. Here, we report a strategy to assemble C3dg into supramolecular nanofibers with excellent compositional control, using β-tail fusion tags. These assemblies were investigated as therapeutic active immunotherapies, which may offer advantages over existing biologics, particularly toward chronic inflammatory diseases. Supramolecular assemblies based on the Q11 peptide system containing β-tail-tagged C3dg, B cell epitopes from TNF, and the universal T cell epitope PADRE raised strong antibody responses against both TNF and C3dg, and prophylactic immunization with these materials significantly improved protection in a lethal TNF-mediated inflammation model. Additionally, in a murine model of psoriasis induced by imiquimod, the C3dg-adjuvanted nanofiber vaccine performed as well as anti-TNF monoclonal antibodies. Nanofibers containing only β-tail-C3dg and lacking the TNF B cell epitope also showed improvements in both models, suggesting that supramolecular C3dg, by itself, played an important therapeutic role. We observed that immunization with β-tail-C3dg caused the expansion of an autoreactive C3dg-specific T cell population, which may act to dampen the immune response, preventing excessive inflammation. These findings indicate that molecular assemblies displaying C3dg warrant further development as active immunotherapies.
Collapse
|
10
|
Avni D, Harikumar KB, Sanyal AJ, Spiegel S. Deletion or inhibition of SphK1 mitigates fulminant hepatic failure by suppressing TNFα-dependent inflammation and apoptosis. FASEB J 2021; 35:e21415. [PMID: 33566377 PMCID: PMC8491138 DOI: 10.1096/fj.202002540r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 12/13/2022]
Abstract
Acute liver failure (ALF) causes severe liver dysfunction that can lead to multi-organ failure and death. Previous studies suggest that sphingosine kinase 1 (SphK1) protects against hepatocyte injury, yet not much is still known about its involvement in ALF. This study examines the role of SphK1 in D-galactosamine (GalN)/lipopolysaccharide (LPS)-induced ALF, which is a well-established experimental mouse model that mimics the fulminant hepatitis. Here we report that deletion of SphK1, but not SphK2, dramatically decreased GalN/LPS-induced liver damage, hepatic apoptosis, serum alanine aminotransferase levels, and mortality rate compared to wild-type mice. Whereas GalN/LPS treatment-induced hepatic activation of NF-κB and JNK in wild-type and SphK2-/- mice, these signaling pathways were reduced in SphK1-/- mice. Moreover, repression of ALF in SphK1-/- mice correlated with decreased expression of the pro-inflammatory cytokine TNFα. Adoptive transfer experiments indicated that SphK1 in bone marrow-derived infiltrating immune cells but not in host liver-resident cells, contribute to the development of ALF. Interestingly, LPS-induced TNFα production was drastically suppressed in SphK1-deleted macrophages, whereas IL-10 expression was markedly enhanced, suggesting a switch to the anti-inflammatory phenotype. Finally, treatment with a specific SphK1 inhibitor ameliorated inflammation and protected mice from ALF. Our findings suggest that SphK1 regulates TNFα secretion from macrophages and inhibition or deletion of SphK1 mitigated ALF. Thus, a potent inhibitor of SphK1 could potentially be a therapeutic agent for fulminant hepatitis.
Collapse
Affiliation(s)
- Dorit Avni
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Kuzhuvelil B. Harikumar
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Arun J. Sanyal
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| |
Collapse
|
11
|
Protective effect of diosgenin on LPS/D-Gal-induced acute liver failure in C57BL/6 mice. Microb Pathog 2020; 146:104243. [DOI: 10.1016/j.micpath.2020.104243] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 02/20/2020] [Accepted: 04/30/2020] [Indexed: 12/28/2022]
|
12
|
Zagórska A, Través PG, Jiménez-García L, Strickland JD, Oh J, Tapia FJ, Mayoral R, Burrola P, Copple BL, Lemke G. Differential regulation of hepatic physiology and injury by the TAM receptors Axl and Mer. Life Sci Alliance 2020; 3:e202000694. [PMID: 32571802 PMCID: PMC7335405 DOI: 10.26508/lsa.202000694] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 05/28/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies have implicated the TAM receptor tyrosine kinase (RTK) Mer in liver disease, yet our understanding of the role that Mer and its related RTKs Tyro3 and Axl play in liver homeostasis and the response to acute injury is limited. We find that Mer and Axl are most prominently expressed in hepatic Kupffer and endothelial cells and that as mice lacking these RTKs age, they develop profound liver disease characterized by apoptotic cell accumulation and immune activation. We further find that Mer is critical to the phagocytosis of apoptotic hepatocytes generated in settings of acute hepatic injury, and that Mer and Axl act in concert to inhibit cytokine production in these settings. In contrast, we find that Axl is uniquely important in mitigating liver damage during acetaminophen intoxication. Although Mer and Axl are protective in acute injury models, we find that Axl exacerbates fibrosis in a model of chronic injury. These divergent effects have important implications for the design and implementation of TAM-directed therapeutics that might target these RTKs in the liver.
Collapse
Affiliation(s)
- Anna Zagórska
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Paqui G Través
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Madrid, Spain
| | | | - Jenna D Strickland
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, USA
| | - Joanne Oh
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Francisco J Tapia
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Rafael Mayoral
- Division of Endocrinology & Metabolism, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Burrola
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
| | - Bryan L Copple
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, USA
| | - Greg Lemke
- Molecular Neurobiology Laboratory, The Salk Institute, La Jolla, CA, USA
- Immunobiology and Microbial Pathogenesis Laboratory, The Salk Institute, La Jolla, CA, USA
| |
Collapse
|
13
|
Brito H, Marques V, Afonso MB, Brown DG, Börjesson U, Selmi N, Smith DM, Roberts IO, Fitzek M, Aniceto N, Guedes RC, Moreira R, Rodrigues CMP. Phenotypic high-throughput screening platform identifies novel chemotypes for necroptosis inhibition. Cell Death Discov 2020; 6:6. [PMID: 32123582 PMCID: PMC7026080 DOI: 10.1038/s41420-020-0240-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 02/07/2023] Open
Abstract
Regulated necrosis or necroptosis, mediated by receptor-interacting kinase 1 (RIPK1), RIPK3 and pseudokinase mixed lineage kinase domain-like protein (MLKL), contributes to the pathogenesis of inflammatory, infectious and degenerative diseases. Recently identified necroptosis inhibitors display moderate specificity, suboptimal pharmacokinetics, off-target effects and toxicity, preventing these molecules from reaching the clinic. Here, we developed a cell-based high-throughput screening (HTS) cascade for the identification of small-molecule inhibitors of necroptosis. From the initial library of over 250,000 compounds, the primary screening phase identified 356 compounds that strongly inhibited TNF-α-induced necroptosis, but not apoptosis, in human and murine cell systems, with EC50 < 6.7 μM. From these, 251 compounds were tested for RIPK1 and/or RIPK3 kinase inhibitory activity; some were active and several have novel mechanisms of action. Based on specific chemical descriptors, 110 compounds proceeded into the secondary screening cascade, which then identified seven compounds with maximum ability to reduce MLKL activation, IC50 >100 μM, EC50 2.5-11.5 μM under long-term necroptosis execution in murine fibroblast L929 cells, and full protection from ATP depletion and membrane leakage in human and murine cells. As a proof of concept, compound SN-6109, with binding mode to RIPK1 similar to that of necrostatin-1, confirmed RIPK1 inhibitory activity and appropriate pharmacokinetic properties. SN-6109 was further tested in mice, showing efficacy against TNF-α-induced systemic inflammatory response syndrome. In conclusion, a phenotypic-driven HTS cascade promptly identified robust necroptosis inhibitors with in vivo activity, currently undergoing further medicinal chemistry optimization. Notably, the novel hits highlight the opportunity to identify new molecular mechanisms of action in necroptosis.
Collapse
Affiliation(s)
- Hugo Brito
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Vanda Marques
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Marta B. Afonso
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Dean G. Brown
- Hit Discovery, Discovery Sciences, R&D Biopharmaceuticals, AstraZeneca, Boston, MA 02451 USA
| | - Ulf Börjesson
- Hit Discovery, Discovery Sciences, R&D Biopharmaceuticals, AstraZeneca, Gothenburg, 431 83 Sweden
| | - Nidhal Selmi
- Hit Discovery, Discovery Sciences, R&D Biopharmaceuticals, AstraZeneca, Gothenburg, 431 83 Sweden
| | - David M. Smith
- Emerging Innovations Unit, Discovery Sciences, R&D, AstraZeneca, Cambridge, CB4 0WG UK
| | - Ieuan O. Roberts
- Hit Discovery, Discovery Sciences, R&D Biopharmaceuticals, AstraZeneca, Cambridge, CB4 0WG UK
| | - Martina Fitzek
- Hit Discovery, Discovery Sciences, R&D Biopharmaceuticals, AstraZeneca, Alderley Park, Macclesfield, SK10 4TG UK
| | - Natália Aniceto
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Rita C. Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Rui Moreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Cecília M. P. Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
14
|
Haderski GJ, Kandar BM, Brackett CM, Toshkov IM, Johnson CP, Paszkiewicz GM, Natarajan V, Gleiberman AS, Gudkov AV, Burdelya LG. TLR5 agonist entolimod reduces the adverse toxicity of TNF while preserving its antitumor effects. PLoS One 2020; 15:e0227940. [PMID: 32027657 PMCID: PMC7004342 DOI: 10.1371/journal.pone.0227940] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 01/02/2020] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor alpha (TNF) is capable of inducing regression of solid tumors. However, TNF released in response to Toll-like receptor 4 (TLR4) activation by bacterial lipopolysaccharide (LPS) is the key mediator of cytokine storm and septic shock that can cause severe tissue damage limiting anticancer applications of this cytokine. In our previous studies, we demonstrated that activation of another Toll-like receptor, TLR5, could protect from tissue damage caused by a variety of stresses including radiation, chemotherapy, Fas-activating antibody and ischemia-reperfusion. In this study, we tested whether entolimod could counteract TNF-induced toxicity in mouse models. We found that entolimod pretreatment effectively protects livers and lungs from LPS- and TNF-induced toxicity and prevents mortality caused by combining either of these agents with the sensitizer, D-galactosamine. While LPS and TNF induced significant activation of apoptotic caspase 3/7, lipid tissue peroxidation and serum ALT accumulation in mice without entolimod treatment, these indicators of toxicity were reduced by entolimod pretreatment to the levels of untreated control mice. Entolimod was effective when injected 0.5–48 hours prior to, but not when injected simultaneously or after LPS or TNF. Using chimeric mice with hematopoiesis differing in its TLR5 status from the rest of tissues, we showed that this protective activity was dependent on TLR5 expression by non-hematopoietic cells. Gene expression analysis identified multiple genes upregulated by entolimod in the liver and cultured hepatocytes as possible mediators of its protective activity. Entolimod did not interfere with the antitumor activity of TNF in mouse hepatocellular and colorectal tumor models. These results support further development of TLR5 agonists to increase tissue resistance to cytotoxic cytokines, reduce the risk of septic shock and enable safe systemic application of TNF as an anticancer therapy.
Collapse
Affiliation(s)
- Gary J. Haderski
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Bojidar M. Kandar
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Craig M. Brackett
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Ilia M. Toshkov
- Genome Protection, Inc., Buffalo, New York, United States of America
| | - Christopher P. Johnson
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Geraldine M. Paszkiewicz
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | - Venkatesh Natarajan
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
| | | | - Andrei V. Gudkov
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
- * E-mail: (LGB); (AVG)
| | - Lyudmila G. Burdelya
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, United States America
- * E-mail: (LGB); (AVG)
| |
Collapse
|
15
|
|
16
|
Wu X, Lei C, Xia T, Zhong X, Yang Q, Shu HB. Regulation of TRIF-mediated innate immune response by K27-linked polyubiquitination and deubiquitination. Nat Commun 2019; 10:4115. [PMID: 31511519 PMCID: PMC6739404 DOI: 10.1038/s41467-019-12145-1] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/21/2019] [Indexed: 01/12/2023] Open
Abstract
TIR domain-containing adaptor inducing interferon-β (TRIF) is an essential adaptor protein required for innate immune responses mediated by Toll-like receptor (TLR) 3- and TLR4. Here we identify USP19 as a negative regulator of TLR3/4-mediated signaling. USP19 deficiency increases the production of type I interferons (IFN) and proinflammatory cytokines induced by poly(I:C) or LPS in vitro and in vivo. Usp19-/- mice have more serious inflammation after poly(I:C) or LPS treatment, and are more susceptible to inflammatory damages and death following Salmonella typhimurium infection. Mechanistically, USP19 interacts with TRIF and catalyzes the removal of TRIF K27-linked polyubiquitin moieties, thereby impairing the recruitment of TRIF to TLR3/4. In addition, the RING E3 ubiquitin ligase complex Cullin-3-Rbx1-KCTD10 catalyzes K27-linked polyubiquitination of TRIF at K523, and deficiency of this complex inhibits TLR3/4-mediated innate immune signaling. Our findings thus reveal TRIF K27-linked polyubiquitination and deubiquitination as a critical regulatory mechanism of TLR3/4-mediated innate immune responses. TRIF is an important adaptor protein for mediating Toll-like receptor (TLR) 3 and TLR4 signaling. Here the authors show that the deubiquitinating enzymes USP19, as well as the E3 ubiquitin ligase complex Cullin-3-Rbx1-KCTD10, modulates TRIF K523 ubiquitination and thereby TRIF recruitment to TLR3/4 to control innate immunity.
Collapse
Affiliation(s)
- Xin Wu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China.,Department of Cell Biology, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Caoqi Lei
- Department of Cell Biology, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Tian Xia
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China.,Department of Cell Biology, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Xuan Zhong
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China.,Department of Cell Biology, College of Life Sciences, Wuhan University, 430072, Wuhan, China
| | - Qing Yang
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430071, Wuhan, China. .,Department of Cell Biology, College of Life Sciences, Wuhan University, 430072, Wuhan, China.
| |
Collapse
|
17
|
Wang SY, Li M, Miao LY, Wu S, Tong YF, Zhang WX, Zhang YY, Sun H. Protective effects of a novel water-soluble biphenyl compound WLP-S-14 against acute-on-chronic liver failure in rats. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2019; 21:928-938. [PMID: 31111726 DOI: 10.1080/10286020.2019.1585822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 02/19/2019] [Accepted: 02/19/2019] [Indexed: 06/09/2023]
Abstract
This study investigated the therapeutic effects of a water-soluble biphenyl compound, WLP-S-14, in acute-on-chronic liver failure (ACLF). Wistar rats were injected intraperitoneally with porcine serum twice a week for 8 weeks prior to administration of 600 mg/kg D-galactosamine and 50 μg/kg lipopolysaccharide to induce ACLF. Study groups were treated intravenously with saline or with 100 or 200 mg/kg WLP-S-14. WLP-S-14 ameliorated ACLF with significant reductions in the mortality rate and transaminase levels, indicating improved liver function. The mechanism underlying these effects may involve decreased levels of tumor necrosis factor-α and interleukin-6, with associated inhibition of apoptotic pathways.
Collapse
Affiliation(s)
- Shao-Yuan Wang
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| | - Mei Li
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| | - Lu-Yang Miao
- b School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Song Wu
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| | - Yuan-Feng Tong
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| | - Wen-Xuan Zhang
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| | - Yu-Yang Zhang
- b School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University , Shenyang 110016 , China
| | - Hua Sun
- a State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Material Medica, Chinese Academy of Medical Science & Peking Union Medical College , Beijing 100050 , China
| |
Collapse
|
18
|
Elufioye TO, Habtemariam S. Hepatoprotective effects of rosmarinic acid: Insight into its mechanisms of action. Biomed Pharmacother 2019; 112:108600. [PMID: 30780110 DOI: 10.1016/j.biopha.2019.108600] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 01/11/2019] [Accepted: 01/18/2019] [Indexed: 02/06/2023] Open
Abstract
Liver diseases such as hepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma are one of the major health challenges in the world and many conditions such as inadequate nutrition, viral infection, ethanol and drug abuse, xenobiotic exposure, and metabolic diseases have been implicated in the development and progression of liver diseases. Several factors including lipid peroxidation, production of reactive oxygen species (ROS), peroxynitrite formation, complement factors and proinflammatory mediators, such as cytokines and chemokines, are involved in hepatic diseases. Rosmarinic acid (RA) is a natural phenolic compound found mainly in the family Lamiaceae consisting of several medicinal plants, herbs and spices. Several biological activities have been reported for RA and these include antioxidant properties as a ROS scavenger and lipid peroxidation inhibitor, anti-inflammatory, neuroprotective and antiangiogenic among others. This review is aimed at discussing the effects of RA on the liver, highlighting its hepatoprotective potential and the underlying mechanisms.
Collapse
Affiliation(s)
- Taiwo O Elufioye
- Department of Pharmacognosy, Faculty of Pharmacy, University of Ibadan, Nigeria.
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories & Herbal Analysis Services, University of Greenwich, Chatham, Maritime Kent, ME4 4TB, UK
| |
Collapse
|
19
|
Li Y, Lv L, Ye J, Fang D, Shi D, Wu W, Wang Q, Wu J, Yang L, Bian X, Jiang X, Jiang H, Yan R, Peng C, Li L. Bifidobacterium adolescentis CGMCC 15058 alleviates liver injury, enhances the intestinal barrier and modifies the gut microbiota in D-galactosamine-treated rats. Appl Microbiol Biotechnol 2018; 103:375-393. [PMID: 30345482 DOI: 10.1007/s00253-018-9454-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
Acute liver failure is a drastic, unpredictable clinical syndrome with high mortality. Various preventive and adjuvant therapies based on modulating the gut flora have been proposed for hepatic injury. We aimed to explore the preventive and therapeutic effects of Bifidobacterium adolescentis CGMCC15058 on rat liver failure, as well as the potential microecological and immunological mechanisms of those effects. B. adolescentis CGMCC15058 (3 × 109 CFU), isolated from healthy human stool, was gavaged to Sprague-Dawley rats for 14 days. Acute liver injury was induced on the 15th day by intraperitoneal injection of D-galactosamine. After 24 h, liver and terminal ileum histology, liver function, plasma cytokines, bacterial translocation and gut microbiota composition were assessed. We found that pretreatment with B. adolescentis significantly relieved elevated serum levels of alanine aminotransferase (ALT), total bile acid and lipopolysaccharide-binding protein and enhanced the expression of mucin 4 and the tight junction protein zonula occludens-1. B. adolescentis exhibited anti-inflammatory properties as indicated by decreased levels of mTOR and the inflammatory cytokines TNF-α and IL-6, as well as elevated levels of the anti-inflammatory cytokine interleukins-10 in the liver. Similar anti-inflammatory signs were also found in plasma. B. adolescentis significantly altered the microbial community, depleting the common pathogenic taxon Proteus and markedly enriching the taxa Coriobacteriaceae, Bacteroidales and Allobaculum, which are involved in regulating the metabolism of lipids and aromatic amino acids. Our findings not only suggest B. adolescentis acts as a prospective probiotic against liver failure but also provide new insights into the prevention and treatment of liver disease.
Collapse
Affiliation(s)
- Yating Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jianzhong Ye
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Daiqiong Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ding Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Wenrui Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Qing Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Jingjing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Liya Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xiaoyuan Bian
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Xianwan Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Huiyong Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Ren Yan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Conggao Peng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China.,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 31003, People's Republic of China. .,Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, China.
| |
Collapse
|
20
|
Choiniere J, Lin MJ, Wang L, Wu J. Deficiency of pyruvate dehydrogenase kinase 4 sensitizes mouse liver to diethylnitrosamine and arsenic toxicity through inducing apoptosis. LIVER RESEARCH 2018; 2:100-107. [PMID: 31815032 PMCID: PMC6896988 DOI: 10.1016/j.livres.2018.05.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIM Pyruvate dehydrogenase kinase 4 (PDK4) is a metabolism switch that regulates glucose oxidation and the tricarboxylic acid cycle (TCA cycle) in the mitochondria. Liver detoxifies xenobiotics and is constantly challenged by various injuries. This study aims at understanding how the loss of the metabolism regulator PDK4 contributes to liver injuries. METHODS Wild-type (WT) and Pdk4 knockout (Pdk4 -/-) mice of different ages were examined for spontaneous hepatic apoptosis. Juvenile or adult mice of two genotypes were insulted by diethylnitrosamine (DEN), arsenic, galactosamine (GalN)/lipopolysaccharide (LPS), anti-CD95 (Jo2) antibody or carbon tetrachloride (CCl4). Liver injury was monitored by blood biochemistry test. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, poly (ADP-ribose) polymerase (PARP) cleavage, and caspase activity assay. Inflammatory response was determined by nuclear factor (NF)-κB activation and the activation of NF-κB target genes. Primary hepatocytes were isolated and cell viability was evaluated by MTS assay. RESULTS We showed that systematic Pdk4 -/- in mice resulted in age-dependent spontaneous hepatic apoptosis. PDK4-deficiency increased the toxicity of DEN in juvenile mice, which correlated with a lethal consequence and massive hepatic apoptosis. Similarly, chronic arsenic administration induced more severe hepatic apoptosis in Pdk4 -/- mice compared to WT control mice. An aggravated hepatic NF-κB mediated-inflammatory response was observed in Pdk4 -/- mice livers. In vitro, Pdk4-deficient primary hepatocytes were more vulnerable to DEN and arsenic challenges and displayed higher caspase activity than wild type cells. Notably, hepatic PDK4 mRNA level was remarkably reduced during acute liver failure induced by GalN/LPS or Jo2 antibody. The diminished PDK4 expression was also observed in CCl4-induced acute liver injury. CONCLUSIONS PDK4 may contribute to the protection from apoptotic injury in mouse liver.
Collapse
Affiliation(s)
- Jonathan Choiniere
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Matthew Junda Lin
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| | - Li Wang
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
- Veterans Affairs Connecticut Healthcare System, West Haven, CT, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, USA
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianguo Wu
- Department of Physiology and Neurobiology, Institute for Systems Genomics, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
21
|
Liu W, Jing ZT, Wu SX, He Y, Lin YT, Chen WN, Lin XJ, Lin X. A Novel AKT Activator, SC79, Prevents Acute Hepatic Failure Induced by Fas-Mediated Apoptosis of Hepatocytes. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1171-1182. [PMID: 29673487 DOI: 10.1016/j.ajpath.2018.01.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/16/2017] [Accepted: 01/08/2018] [Indexed: 01/18/2023]
Abstract
Acute liver failure is a serious clinical problem of which the underlying pathogenesis remains unclear and for which effective therapies are lacking. The Fas receptor/ligand system, which is negatively regulated by AKT, is known to play a prominent role in hepatocytic cell death. We hypothesized that AKT activation may represent a strategy to alleviate Fas-induced fulminant liver failure. We report here that a novel AKT activator, SC79, protects hepatocytes from apoptosis induced by agonistic anti-Fas antibody CH11 (for humans) or Jo2 (for mice) and significantly prolongs the survival of mice given a lethal dose of Jo2. Under Fas-signaling stimulation, SC79 inhibited Fas aggregation, prevented the recruitment of the adaptor molecule Fas-associated death domain (FADD) and procaspase-8 [or FADD-like IL-1β-converting enzyme (FLICE)] into the death-inducing signaling complex (DISC), but SC79 enhanced the recruitment of the long and short isoforms of cellular FLICE-inhibitory protein at the DISC. All of the SC79-induced hepatoprotective and DISC-interruptive effects were confirmed to have been reversed by the Akt inhibitor LY294002. These results strongly indicate that SC79 protects hepatocytes from Fas-induced fatal hepatic apoptosis. The potent alleviation of Fas-mediated hepatotoxicity by the relatively safe drug SC79 highlights the potential of our findings for immediate hepatoprotective translation.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zhen-Tang Jing
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Shu-Xiang Wu
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yun He
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yan-Ting Lin
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Wan-Nan Chen
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China
| | - Xin-Jian Lin
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Xu Lin
- Key Laboratory of the Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China; Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
22
|
Li JJ, Li Z, Gu LJ, Choi KJ, Kim DS, Kim HK, Sung CK. The promotion of hair regrowth by topical application of a Perilla frutescens extract through increased cell viability and antagonism of testosterone and dihydrotestosterone. J Nat Med 2018; 72:96-105. [PMID: 28905175 DOI: 10.1007/s11418-017-1116-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 07/19/2017] [Indexed: 11/25/2022]
Abstract
This study investigated the potential hair regrowth effects associated with a plant extract of Perilla frutescens, which was selected due to its putative hair regrowth activity. Extracts were prepared from dried P. frutescens suspended in distilled water, where the resultant aqueous suspension was fractionated sequentially using hexane, ethyl acetate, n-butanol, and distilled water. We observed that the n-butanol fraction resulted in the highest hair regrowth activity. The n-butanol soluble fraction of P. frutescens extract (BFPE) was further separated using AB-8 macroporous resin and silica gel chromatography to obtain rosmarinic acid (RA), which demonstrated effective hair growth regeneration potential. BFPE also showed in vivo anti-androgenic activity following the use of a hair growth assay in testosterone-sensitive male C57Bl/6NCrSlc mice. Furthermore, the effects of cell viability promotion were investigated following an in vitro analysis in primary hair follicle fibroblast cells (PHFCs) treated with RA. The results suggested that RA was the active compound in P. frutescens that triggers hair growth, and RA could be a potential therapeutic agent for the promotion of hair growth and prevention of androgenetic alopecia (AGA).
Collapse
Affiliation(s)
- Jing-Jie Li
- Mibyeong Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Zheng Li
- Intelligent Synthetic Biology Center, KAIST, Daejeon, 34141, Republic of Korea
| | - Li-Juan Gu
- Department of Food Science and Technology, College of Agriculture and Biotechnology, Chungnam National University, 220 Gung-dong, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Kang-Ju Choi
- Department of Food Science and Technology, College of Agriculture and Biotechnology, Chungnam National University, 220 Gung-dong, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Dong-Seon Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Ho-Kyoung Kim
- Mibyeong Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea.
| | - Chang-Keun Sung
- Department of Food Science and Technology, College of Agriculture and Biotechnology, Chungnam National University, 220 Gung-dong, Yusung-gu, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
23
|
Metal transporter Slc39a10 regulates susceptibility to inflammatory stimuli by controlling macrophage survival. Proc Natl Acad Sci U S A 2017; 114:12940-12945. [PMID: 29180421 PMCID: PMC5724256 DOI: 10.1073/pnas.1708018114] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Zn is essential for maintaining the integrity of the immune system, and Zn homeostasis is tightly regulated by two families of ion transporters, SLC39A and SLC30A. Worldwide, an estimated two billion people have Zn deficiency, a condition that can impair immune function and increase susceptibility to a variety of infections. Despite their important roles in health and disease, the molecular mechanisms that underlie Zn transport and Zn homeostasis in macrophages are poorly understood. Here, we report that SLC39A10 plays an essential role in Zn homeostasis in macrophages, regulating the immune response following inflammatory stimuli. Specifically, we identified a role for SLC39A10 in regulating the survival of macrophages via a Zn/p53-dependent axis during the inflammatory response. Zn plays a key role in controlling macrophage function during an inflammatory event. Cellular Zn homeostasis is regulated by two families of metal transporters, the SLC39A family of importers and the SLC30A family of exporters; however, the precise role of these transporters in maintaining macrophage function is poorly understood. Using macrophage-specific Slc39a10-knockout (Slc39a10fl/fl;LysM-Cre+) mice, we found that Slc39a10 plays an essential role in macrophage survival by mediating Zn homeostasis in response to LPS stimulation. Compared with Slc39a10fl/fl mice, Slc39a10fl/fl;LysM-Cre+ mice had significantly lower mortality following LPS stimulation as well as reduced liver damage and lower levels of circulating inflammatory cytokines. Moreover, reduced intracellular Zn concentration in Slc39a10fl/fl;LysM-Cre+ macrophages led to the stabilization of p53, which increased apoptosis upon LPS stimulation. Concomitant knockout of p53 largely rescued the phenotype of Slc39a10fl/fl;LysM-Cre+ mice. Finally, the phenotype in Slc39a10fl/fl;LysM-Cre+ mice was mimicked in wild-type mice using the Zn chelator TPEN and was reversed with Zn supplementation. Taken together, these results suggest that Slc39a10 plays a role in promoting the survival of macrophages through a Zn/p53-dependent axis in response to inflammatory stimuli.
Collapse
|
24
|
Raghuvanshi R, Chaudhari A, Kumar GN. 2-Ketogluconic acid and pyrroloquinoline quinone secreting probiotic Escherichia coli Nissle 1917 as a dietary strategy against heavy metal induced damage in rats. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.08.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
25
|
Mora-Solano C, Wen Y, Han H, Chen J, Chong AS, Miller ML, Pompano RR, Collier JH. Active immunotherapy for TNF-mediated inflammation using self-assembled peptide nanofibers. Biomaterials 2017; 149:1-11. [PMID: 28982051 DOI: 10.1016/j.biomaterials.2017.09.031] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/18/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022]
Abstract
Active immunotherapies raising antibody responses against autologous targets are receiving increasing interest as alternatives to the administration of manufactured antibodies. The challenge in such an approach is generating protective and adjustable levels of therapeutic antibodies while at the same time avoiding strong T cell responses that could lead to autoimmune reactions. Here we demonstrate the design of an active immunotherapy against TNF-mediated inflammation using short synthetic peptides that assemble into supramolecular peptide nanofibers. Immunization with these materials, without additional adjuvants, was able to break B cell tolerance and raise protective antibody responses against autologous TNF in mice. The strength of the anti-TNF antibody response could be tuned by adjusting the epitope content in the nanofibers, and the T-cell response was focused on exogenous and non-autoreactive T-cell epitopes. Immunization with unadjuvanted peptide nanofibers was therapeutic in a lethal model of acute inflammation induced by intraperitoneally delivered lipopolysaccharide, whereas formulations adjuvanted with CpG showed comparatively poorer protection that correlated with a more Th1-polarized response. Additionally, immunization with peptide nanofibers did not diminish the ability of mice to clear infections of Listeria monocytogenes. Collectively this work suggests that synthetic self-assembled peptides can be attractive platforms for active immunotherapies against autologous targets.
Collapse
Affiliation(s)
- Carolina Mora-Solano
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Yi Wen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Huifang Han
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Jianjun Chen
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Anita S Chong
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States
| | - Michelle L Miller
- Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22904, United States
| | - Joel H Collier
- Department of Surgery, University of Chicago, Chicago, IL, 60637, United States; Molecular Pathogenesis Program, University of Chicago, Chicago, IL, 60637, United States.
| |
Collapse
|
26
|
Galanos C, Gumenscheimer M, Mühlradt P, Jirillo E, Freudenberg M. MALP-2, a Mycoplasma lipopeptide with classical endotoxic properties: end of an era of LPS monopoly? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060061001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although some activities of LPS are shared by other bacterial components, for half a century LPS has been regarded as unique in displaying many pathophysiological activities. Here we report on a synthetic lipopeptide, MALP-2 from Mycoplasma fermentans , which expresses potent endotoxin-like activity and whose lethal toxicity is comparable to that of LPS. With the exception of the Limulus lysate gelation test, in which MALP-2 was approximately 1000-fold less active than LPS, the synthetic lipopeptide induced all activities tested for, and in most cases to an extent comparable to that of LPS. Unlike LPS, the biological activities of MALP-2 were expressed both in LPSresponder and in LPS-non-responder mice (BALB/c/l, C57BL10/ScCr), indicating that MALP-2 signaling, unlike that of LPS, is not transduced via the Toll-like receptor (Tlr) 4 protein. MALP-2 expressed no toxicity in normal or sensitized Tlr2 knockout (Tlr2— /—) mice indicating that its toxic activity is induced via Tlr2 signaling. The phenomenology of the lethal shock induced by MALP-2 in normal or sensitized mice, i.e. the kinetics of its development and symptoms of illness exhibited by the treated animals, was very reminiscent of the lethal shock induced by LPS.
Collapse
Affiliation(s)
- C. Galanos
- Max-Planck Institut für Immunbiologie, Freiburg, Germany
| | | | - P.F. Mühlradt
- Immunobiology Research Group, GBF, Braunschweig, Germany
| | - E. Jirillo
- Department of Immunology, Faculty of Medicina, Policlinico University, Bari, Italy
| | | |
Collapse
|
27
|
Hasunuma R, Maruyama H, Takimoto H, Ryll R, Tanaka S, Kumazawa Y. Does high mobility group 1 protein function as a late mediator for LPS- or TNF-induced shock in galactosamine-sensitized mice? ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519020080050201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
The role of high mobility group-1 protein (HMG-1) in LPS- and TNF-a-induced lethal shock in galactosamine (GalN)-sensitized mice was investigated. No detectable HMG-1 levels were observed by immunoblotting analysis in plasma from untreated or GalN-sensitized BALB/c mice 5 h after LPS injection, although significant levels of HMG-1 were detected in plasma 6 h after the challenge. All GalN-sensitized BALB/c but not BALB/lpsd mice succumbed by 6 h after LPS injection. When GalN-sensitized mice were injected with TNF-α, the presence of HMG-1 was seen at 5.5 h in plasma of BALB/c mice and at 6 h in BALB/lpsd mice, although almost all GalN-sensitized BALB/c mice died by 6 h after challenge. The time-dependent phenomenon correlated with elevated serum aspartate aminotransferase (AST) levels and the appearance of apoptotic cells in livers. Administration of pooled plasma, equivalent to approximately 200 μg recombinant murine HMG-1, taken from mice on the verge of near death, did not result in induction of lethal shock in GalN-sensitized mice. Taken together with the late appearance of HMG-1 in moribund mice, these data suggest that HMG-1 does not decisively contribute to lethality in the GalN sensitization model.
Collapse
Affiliation(s)
- Ryoichi Hasunuma
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan, -u.ac.jp
| | - Hiroko Maruyama
- Department of Pathology, School of Allied Health Sciences, Kitasato University, Sagamihara, Japan
| | - Hiroaki Takimoto
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - Roland Ryll
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | | | - Yoshio Kumazawa
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| |
Collapse
|
28
|
Yasuda S, Nagaki M, Moriwaki H. Staphylococcal enterotoxin B induces hepatic injury and lethal shock in endotoxin-resistant C3H/HeJ mice despite a deficient macrophage response. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519020080040201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Bacterial toxins, including endotoxin/LPS as well as superantigens, are major causative agents of multi-organ failure associated with sepsis and liver disease. However, the precise mechanisms initiating cell activation by the toxins have not been clarified. We compared lethal shock and cytokine production in response to LPS with responses to the superantigen staphylococcal enterotoxin B (SEB) in both LPS-responsive C3H/HeN mice and LPS-hyporesponsive C3H/HeJ mice treated with D-galactosamine (GalN). LPS was not lethal and did not induce production of TNF-α in C3H/HeJ mice. In contrast, SEB produced lethal shock associated with liver failure and induced cytokines such as TNF-α, IFN-γ, and IL-2 in both C3H/HeN and C3H/HeJ mice. Peritoneal macrophages from C3H/HeJ mice did not produce TNF-α in vitro in response to SEB or LPS. However, no significant difference was observed in production of TNF-α in response to stimulation in vitro by SEB between C3H/HeN and C3H/HeJ splenic lymphocytes. We have demonstrated that SEB causes lethal toxicity associated with liver injury in LPS-hyporesponsive C3H/HeJ mice and that as the underlying mechanism, the normal T-cell function in these mice still maintained the sensitivity to SEB since the genetic defect of C3H/HeJ mice unresponsive to LPS and SEB is restricted in macrophages/monocytes and does not extend to T cells.
Collapse
Affiliation(s)
- Shigeo Yasuda
- First Department of Internal Medicine, Gifu University School of Medicine, Gifu, Japan
| | - Masahito Nagaki
- First Department of Internal Medicine, Gifu University School of Medicine, Gifu, Japan, -u.ac.jp
| | - Hisataka Moriwaki
- First Department of Internal Medicine, Gifu University School of Medicine, Gifu, Japan
| |
Collapse
|
29
|
Bucklin S, Russell S, Morrison D. Augmentation of anti-cytokine immunotherapy by combining neutralizing monoclonal antibodies to interferon-γ and the interferon-γ receptor: protection in endotoxin shock. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199400100108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
We have employed neutralizing monoclonal antibodies to mouse interferon-γ (IFNγ) and to the receptor for mouse IFNy in studies designed to assess the protective efficacy of each of these monoclonals, administered either separately or in combination, in endotoxin-induced lethality. While pretreatment with either antibody alone, at doses of 200 μg per mouse, provided limited protection (70-50% lethality) in comparison to non-neutralizing antibody controls (100% lethality), the two monoclonal antibodies administered together provided a substantially greater level of protection (17% lethality). Although administration of 100 μg per mouse of either monoclonal alone was not protective (more than 65% lethality), a combination of both antibodies at this dose provided significant protection (19% lethality). In addition, administration of both antibodies 30 min post-endotoxin challenge also demonstrated significant protection in comparison to single antibody immunotherapy. In vitro studies using mouse peritoneal macrophages stimulated with LPS and IFNy have established confirmatory data for a synergistic effect of neutralizing antibody to IFNy and the IFNγ receptor in inhibiting macrophage activation as assessed by production of nitric oxide. These results provide a strong rationale for dual targeting of ligand and receptor in single cytokine immunotherapy.
Collapse
Affiliation(s)
- S.E. Bucklin
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, USA, Department of Pathology and Oncology, and The Cancer Center, University of Kansas Medical Center, Kansas City, USA
| | - S.W. Russell
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, USA, Department of Pathology and Oncology, and The Cancer Center, University of Kansas Medical Center, Kansas City, USA
| | - D.C. Morrison
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, USA, Department of Pathology and Oncology, and The Cancer Center, University of Kansas Medical Center, Kansas City, USA
| |
Collapse
|
30
|
Heumann D, Le Roy D, Glauser MP. Contribution of TNF and of LPS in endotoxemic shock in mice. A reappraisal. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Species of mice have a different sensitivity to LPS. The mechanisms underlying these differences are not clear. We have investigated the contribution of TNF and of LPS in inducing lethality in endotoxemic models using LPSsensitive C57BI/6 and Balb/c mice, OF1 mice with an intermediate sensitivity, and LPS-resistant C3H/HeJ mice. Our observations showed that TNF largely contributed to lethality, since anti-TNF antibodies protected sensitive Balb/c mice from death over a large range of LPS concentrations. OF1 mice were relatively insensitive to LPS and this was associated with a lesser production of TNF. Similarly, OF1 mice had a reduced sensitivity to challenge with recombinant TNF than Balb/c mice. These observations, therefore, indicated that TNF production and sensitivity were determinant for outcome upon LPS challenge. LPS levels were similar in Balb/c, C57BI/6, and OF1 mice, suggesting that a difference in LPS clearance did not contribute to the differences in sensitivity to LPS. The contribution of the combination of both TNF and LPS in endotoxemic shock was investigated with the LPS-resistant C3H/HeJ mice. While these mice were resistant to challenge with either LPS or a low dose of TNF, combination of non lethal concentrations of LPS and TNF induced death. These data, thus, suggest that while TNF is critical for mediating LPS-induced death, the combination of both LPS and TNF act synergistically in contributing to death, particularly in animal species with reduced sensitivity to LPS.
Collapse
Affiliation(s)
- Didier Heumann
- Division of Infectious Diseases, CHUV-Lausanne, Switzerland
| | - Didier Le Roy
- Division of Infectious Diseases, CHUV-Lausanne, Switzerland
| | | |
Collapse
|
31
|
Abstract
HMGB1 is an abundant nuclear and cytoplasmic protein present in mammalian cells. It is traditionally known as a DNA binding protein involved in maintenance of nucleosome structure and regulation of gene transcription. Beyond these intracellular roles, we recently discovered that HMGB1 is released from activated macrophages and functions as a late mediator of lethal endotoxemia. Addition of HMGB1 to macrophage cultures activates cytokine release. When released into the extracellular milieu, HMGB1 causes systemic inflammatory responses including acute lung injury, epithelial barrier dysfunction, and death. Passive immunization with anti-HMGB1 antibodies confers significant protection against lethality induced by LPS administration and sepsis caused by cecal perforation in mice. Truncation of HMGB1 into individual structural domains revealed that the HMGB1 A box, a DNA-binding motif, specifically antagonizes the activity of HMGB1 and rescues mice from lethal sepsis caused by cecal perforation. Thus, strategies that target HMGB1 with specific antibodies or antagonists have potential for treating lethal systemic inflammatory diseases characterized by excessive HMGB1 release.
Collapse
Affiliation(s)
- Huan Yang
- Laboratory of Biomedical Science, North Shore-Long Island Jewish Research Institute, Manhasset, New York, USA,
| | - Haichao Wang
- Laboratory of Biomedical Science, North Shore-Long Island Jewish Research Institute, Manhasset, New York, USA
| | - Christopher J. Czura
- Laboratory of Biomedical Science, North Shore-Long Island Jewish Research Institute, Manhasset, New York, USA
| | - Kevin J. Tracey
- Laboratory of Biomedical Science, North Shore-Long Island Jewish Research Institute, Manhasset, New York, USA
| |
Collapse
|
32
|
Abstract
D-Galactosamine (D-galN) is well established as sensitizing mice and other animals to the lethal effects of TNF, specifically, and by several orders of magnitude. Protection by anti-TNF neutralizing antibody is complete, as is (metabolically-based) protection by uridine. Sensitization occurs regardless of the origin of the released TNF, whether it is released from macrophages and/or T-cells. The same is true for the challenging agent which leads to the release of TNF, whether it is endotoxin, a superantigen, lipoprotein, bacterial DNA, or bacteria, either killed or proliferating. Most studies have utilized endotoxin as the challenging agent, and more than 70 agents have been reported to confer protection against LPS and/or TNF challenge in the model. The model has provided new insight regarding modes of protection, including from dexamethasone, which protects against challenge from LPS but not from challenge by TNF. The D-galN lethality model has also been used to test for synergistic behavior between different bacterial components, and to test for lethality when only small amounts of the challenging agent are available (lipid A chemistry).
Collapse
Affiliation(s)
- Richard Silverstein
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA,
| |
Collapse
|
33
|
Morita H, Hasunuma R, Hoshino M, Fujihara M, Tanaka S, Yamamoto S, Kumazawa Y. Difference in clearance of exogenously administered smooth-form LPS following host responses among normal, sensitized and LPS-tolerant mice. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199700400605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Clearance of exogenously administered Salmonella abortus equi LPS from the circulation following induction of host responses, e.g. release of soluble CD14 (sCD14) and TNFα production, were investigated. The endotoxin unit of administered LPS in plasma was monitored by a combined method. After 1 h, more than 80% of injected LPS disappeared from the circulation of normal mice at all doses except a 100 μg dose, but sCD14 in plasma could not yet be detected by Western blotting. Release of sCD14 reached a peak 9 h after LPS injection. According to pretreatment with either Propionibacterium acnes or silver nitrate, the clearance rates of exogenously added LPS from the circulation were accelerated in comparison with the rate in normal mice, but plasma TNF levels were the opposite. In LPS-tolerant mice, LPS clearance and production of TNF and sCD14 was reduced. Pretreatment with anti-CD14 mAb reduced LPS-induced TNF production but did not influence the clearance rates. Taken together, in vivo, sCD14 may not play a critical role for early LPS clearance.
Collapse
Affiliation(s)
- H. Morita
- Seikagaku Corp., Chuo-ku, Tokyo, Japan
| | - R. Hasunuma
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - M. Hoshino
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan
| | - M. Fujihara
- Department of Biochemistry, School of Allied Health Sciences, Kitasato University, Sagamihara, Japan
| | - S. Tanaka
- Seikagaku Corp., Chuo-ku, Tokyo, Japan
| | - S. Yamamoto
- Department of Pathology, Ohita Medical College, Ohita-gun,Japan
| | - Y. Kumazawa
- Department of Biosciences, School of Science, Kitasato University, Sagamihara, Japan, -u.ac.jp
| |
Collapse
|
34
|
Abstract
The LPS binding protein (LBP) is an acute phase protein and an integral component of innate immunity. In normal plasma, LBP was shown to be involved in the response of monocytes to LPS. LBP was also shown to play a detrimental role in models of endotoxemia in mice. Levels of LBP increase during infection. To determine whether the presence of an intact bacterial flora is a prerequisite for normal LBP function, we analyzed the serum concentrations and functional activity of LBP in conventional and germfree mice. Baseline LBP levels were similar in both types of mice, as was the production of the pro-inflammatory cytokines TNFα and IL-6 in whole blood stimulated with LPS. Moreover, D-galactosamine sensitized germfree and conventional mice exhibited similar susceptibility to LPS. We thus conclude that the presence of a bacterial flora is not required to support normal levels and function of LBP.
Collapse
Affiliation(s)
- D. Le Roy
- Division of Infectious Diseases, CHUV-Lausanne, Switzerland
| | - M.P. Glauser
- Division of Infectious Diseases, CHUV-Lausanne, Switzerland
| | - D. Heumann
- Division of Infectious Diseases, CHUV-Lausanne, Switzerland
| |
Collapse
|
35
|
Tsukushi Y, Kido N, Saeki K, Sugiyama T, Koide N, Mori I, Yoshida T, Yokochi T. Characteristic biological activities of lipopolysaccharides from Sinorhizobium and Mesorhizobium. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100010301] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The biological actions of lipopolysaccharides (LPSs) from Sinorhizobium meliloti, Mesorhizobium loti and Escherichia coli were compared. In biological activities including lethality, production of tumor necrosis factor (TNF)-α and nitric oxide (NO), adjuvant action and Limulus activity, LPS from S. meliloti exhibited stronger actions than LPS from M. loti, but had a weaker action than LPS from E. coli. On the other hand, M. loti LPS showed a higher activity to activate human complement than S. meliloti LPS. Further, there was a significant difference in polymyxin B binding between S. meliloti LPS and M. loti LPS, suggesting a difference in the lipid A structure. LPSs from S. meliloti and M. loti seem to exhibit characteristic biological actions that may be dependent on the difference in the lipid A structure.
Collapse
Affiliation(s)
- Yasuyo Tsukushi
- Department of Natural Science Informatics, School of Informatics and Sciences, Nagoya University, Nagoya, Aichi, Japan, Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Nobuo Kido
- Department of Natural Science Informatics, School of Informatics and Sciences, Nagoya University, Nagoya, Aichi, Japan
| | - Kazuhiko Saeki
- Department of Biology, Osaka University Graduate School of Science, Toyonaka, Osaka, Japan
| | - Tsuyoshi Sugiyama
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Naoki Koide
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Isamu Mori
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Tomoaki Yoshida
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Takashi Yokochi
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan,
| |
Collapse
|
36
|
Freudenberg MA, Kopf M, Galanos C. Lipopolysaccharide-sensitivity of interferon-γ receptor deficient mice. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199600300402] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interteron-γ receptor deficient (IFNγR-/-) and control wild-type (IFNγR+/+) mice were compared for their susceptibility to lipopolysaccharide (LPS) and tumor necrosis factor α (TNFα) before and after treatment with the LPSsensitizing agents D-galactosamine (D-GalN) or Propionibacterium acnes. Untreated and D-GalN-sensitized, IFNγR-/and IFNγR+/+ mice were comparable in their TNFα responses to LPS. Deficient and wild-type mice also exhibited a similar susceptibility to the lethal effects of LPS or TNFα which was enhanced by D-GalN treatment to a comparable extent. After treatment with P. acnes, the LPS and TNFα responsiveness remained unchanged in IFNγR-/- mice while in IFNγR+/+ mice they were enhanced up to 200-fold. These results indicate that IFN-γ mediates the LPS and TNFα hypersensitivity of P. acnes treated mice, while the normal sensitivity to LPS or TNFα in the presence or absence of D-GalN is IFN-γ independent.
Collapse
Affiliation(s)
| | - Manfred Kopf
- Basel Institute for Immunology, Basel, Switzerland
| | - Chris Galanos
- Max-Planck-Institut für lmmunbiologie, Freiburg, Germany
| |
Collapse
|
37
|
Chapes S, Beharka A. Lipopolysaccharide is required for the lethal effects of enterotoxin B after D-galactosamine sensitization. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/096805199500200406] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
We tested the D-galactosamine sensitization model with staphylococcal enterotoxin B (SEB). LPS was required for the lethal effects of SEB in D-galactosamine sensitized mice. Only two (2/62) among the C3HeB/FeJ (H-2k), Balb/c (H-2d) and C57BL/6J (H-2b) mice died in response to SEB in the absence of LPS whereas injection of SEB and minimally lethal concentrations of LPS became highly toxic. Similar to LPS, the lethal effect of SEB was dependent on the mouse strain used. Mouse strains more sensitive to the effects of LPS (Balb/c and C57BL/6J) were also more sensitive to the effects of SEB in comparison to C3H mice when equivalent doses of LPS and SEB were used. Among Balb/c and C3HeB/FeJ but not C57BL/6J mice, SEB (20 μg) potentiated the lethal effects of LPS at low doses (0.1 μg LPS), but had an apparent protective effect at high doses (1 μg LPS). Lastly, there was an inverse correlation between pathogenesis and serum IL-2 concentrations and splenic T cell activation in C3H mice. However, macrophage mobilization did correlate with lethality. Therefore, some questions remain about the mechanisms involved in the D-galactosamine/SEB pathogenesis model. We conclude that when sensitizing mice with D-galactosamine and assessing the lethal effects of SEB, endotoxin contamination must be assessed.
Collapse
Affiliation(s)
- S.K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| | - A.A. Beharka
- Division of Biology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
38
|
Lim YC, Rajabalaya R, Lee SHF, Tennakoon KU, Le QV, Idris A, Zulkipli IN, Keasberry N, David SR. Parasitic Mistletoes of the Genera Scurrula and Viscum: From Bench to Bedside. Molecules 2016; 21:1048. [PMID: 27548121 PMCID: PMC6273404 DOI: 10.3390/molecules21081048] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/31/2016] [Accepted: 08/04/2016] [Indexed: 01/16/2023] Open
Abstract
The mistletoes, stem hemiparasites of Asia and Europe, have been used as medicinal herbs for many years and possess sophisticated systems to obtain nutrients from their host plants. Although knowledge about ethnomedicinal uses of mistletoes is prevalent in Asia, systematic scientific study of these plants is still lacking, unlike its European counterparts. This review aims to evaluate the literature on Scurrula and Viscum mistletoes. Both mistletoes were found to have anticancer, antimicrobial, antioxidant and antihypertensive properties. Plants from the genus Scurrula were found to inhibit cancer growth due to presence of phytoconstituents such as quercetin and fatty acid chains. Similar to plants from the genus Viscum, Scurrula also possesses TNFα activity to strengthen the immune system to combat cancer. In line with its anticancer activity, both mistletoes are rich in antioxidants that confer protection against cancer as well as neurodegeneration. Extracts from plants of both genera showed evidence of vasodilation and thus, antihypertensive effects. Other therapeutic effects such as weight loss, postpartum and gastrointestinal healing from different plants of the genus Scurrula are documented. As the therapeutic effects of plants from Scurrula are still in exploration stage, there is currently no known clinical trial on these plants. However, there are few on-going clinical trials for Viscum album that demonstrate the functionalities of these mistletoes. Future work required for exploring the benefits of these plants and ways to develop both parasitic plants as a source of pharmacological drug are explained in this article.
Collapse
Affiliation(s)
- Ya Chee Lim
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Rajan Rajabalaya
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Shirley Huan Fang Lee
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Kushan U Tennakoon
- Institute for Biodiversity and Environmental Research (IBER), Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Quang-Vuong Le
- Institute for Biodiversity and Environmental Research (IBER), Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
- Biology Faculty, Vinh University, 182 Le Duan, 470000 Vinh City, Nghe An Province, Vietnam.
| | - Adi Idris
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Ihsan N Zulkipli
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Natasha Keasberry
- Faculty of Science, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| | - Sheba R David
- Pengiran Anak Puteri Rashidah Sa'adatul Bolkiah Institute of Health Sciences, Universiti Brunei Darussalam, Jalan Tungku Link, BE 1410 Gadong, Brunei.
| |
Collapse
|
39
|
Yao L, Chen W, Han C, Wu T. Microsomal prostaglandin E synthase-1 protects against Fas-induced liver injury. Am J Physiol Gastrointest Liver Physiol 2016; 310:G1071-80. [PMID: 27102561 PMCID: PMC4935489 DOI: 10.1152/ajpgi.00327.2015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 04/17/2016] [Indexed: 01/31/2023]
Abstract
Microsomal prostaglandin E synthase-1 (mPGES-1) is the terminal enzyme for the synthesis of prostaglandin E2 (PGE2), a proproliferative and antiapoptotic lipid molecule important for tissue regeneration and injury repair. In this study, we developed transgenic (Tg) mice with targeted expression of mPGES-1 in the liver to assess Fas-induced hepatocyte apoptosis and acute liver injury. Compared with wild-type (WT) mice, the mPGES-1 Tg mice showed less liver hemorrhage, lower serum alanine transaminase (ALT) and aspartate transaminase (AST) levels, less hepatic necrosis/apoptosis, and lower level of caspase cascade activation after intraperitoneal injection of the anti-Fas antibody Jo2. Western blotting analysis revealed increased expression and activation of the serine/threonine kinase Akt and associated antiapoptotic molecules in the liver tissues of Jo2-treated mPGES-1 Tg mice. Pretreatment with the mPGES-1 inhibitor (MF63) or the Akt inhibitor (Akt inhibitor V) restored the susceptibility of the mPGES-1 Tg mice to Fas-induced liver injury. Our findings provide novel evidence that mPGES-1 prevents Fas-induced liver injury through activation of Akt and related signaling and suggest that induction of mPGES-1 or treatment with PGE2 may represent important therapeutic strategy for the prevention and treatment of Fas-associated liver injuries.
Collapse
Affiliation(s)
| | | | | | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| |
Collapse
|
40
|
Patterns of Transcriptional Response to 1,25-Dihydroxyvitamin D3 and Bacterial Lipopolysaccharide in Primary Human Monocytes. G3-GENES GENOMES GENETICS 2016; 6:1345-55. [PMID: 26976439 PMCID: PMC4856085 DOI: 10.1534/g3.116.028712] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The active form of vitamin D, 1,25-dihydroxyvitamin D3 (1,25D), plays an important immunomodulatory role, regulating transcription of genes in the innate and adaptive immune system. The present study examines patterns of transcriptome-wide response to 1,25D, and the bacterial lipopolysaccharide (LPS) in primary human monocytes, to elucidate pathways underlying the effects of 1,25D on the immune system. Monocytes obtained from healthy individuals of African-American and European-American ancestry were treated with 1,25D, LPS, or both, simultaneously. The addition of 1,25D during stimulation with LPS induced significant upregulation of genes in the antimicrobial and autophagy pathways, and downregulation of proinflammatory response genes compared to LPS treatment alone. A joint Bayesian analysis enabled clustering of genes into patterns of shared transcriptional response across treatments. The biological pathways enriched within these expression patterns highlighted several mechanisms through which 1,25D could exert its immunomodulatory role. Pathways such as mTOR signaling, EIF2 signaling, IL-8 signaling, and Tec Kinase signaling were enriched among genes with opposite transcriptional responses to 1,25D and LPS, respectively, highlighting the important roles of these pathways in mediating the immunomodulatory activity of 1,25D. Furthermore, a subset of genes with evidence of interethnic differences in transcriptional response was also identified, suggesting that in addition to the well-established interethnic variation in circulating levels of vitamin D, the intensity of transcriptional response to 1,25D and LPS also varies between ethnic groups. We propose that dysregulation of the pathways identified in this study could contribute to immune-mediated disease risk.
Collapse
|
41
|
Abstract
Superantigens secreted by Staphylococcus aureus and Streptococcus pyogenes interact with the T-cell receptor and major histocompatibility class II molecules on antigen-presenting cells to elicit a massive cytokine release and activation of T cells in higher numbers than that seen with ordinary antigens. Because of this unique ability, superantigens have been implicated as etiological agents for many different types of diseases, including toxic shock syndrome, infective endocarditis, pneumonia, and inflammatory skin diseases. This review covers the main animal models that have been developed in order to identify the roles of superantigens in human disease.
Collapse
Affiliation(s)
- Amanda J Brosnahan
- Department of Science, Concordia University - Saint Paul, 1282 Concordia Ave., S-115, St. Paul, MN, 55104, USA.
| |
Collapse
|
42
|
Kučera O, Lotková H, Sobotka O, Červinková Z. The effect of D-galactosamine on lean and steatotic rat hepatocytes in primary culture. Physiol Res 2015; 64:S637-S646. [PMID: 26674289 DOI: 10.33549/physiolres.933225] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The aim of our work was to compare the effect of D-galactosamine (GalN) on primary cultures of lean and steatotic rat hepatocytes isolated from intact and fatty liver, respectively. GalN caused more severe injury to steatotic hepatocytes than to lean cells as documented by lactate dehydrogenase leakage. Necrotic mode of cell death strongly prevails over apoptosis since we did not observe any significant increase in activities of caspase 3, 8 and 9 in any group of hepatocytes treated with GalN. Reactive oxygen species (ROS) formation and lipid peroxidation were elevated in a dose-dependent manner by GalN and were significantly more pronounced in fatty hepatocytes. A decrease in the percentage of hepatocytes with energized mitochondria was observed from 30 mM and 10 mM GalN in lean and steatotic hepatocytes, respectively. Our results undoubtedly indicate that steatotic hepatocytes exert higher sensitivity to the toxic effect of GalN. This sensitivity may be caused by more intensive GalN-induced ROS production and lipid peroxidation and by higher susceptibility of mitochondria to loss of mitochondrial membrane potential in steatotic hepatocytes. In our experimental arrangement, apoptosis does not seem to participate considerably on hepatotoxic action of GalN in either group of hepatocytes.
Collapse
Affiliation(s)
- O Kučera
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University in Prague, Hradec Králové, Czech Republic.
| | | | | | | |
Collapse
|
43
|
Yang P, Zhou W, Li C, Zhang M, Jiang Y, Jiang R, Ba H, Li C, Wang J, Yin B, Gong F, Li Z. Kupffer-cell-expressed transmembrane TNF-α is a major contributor to lipopolysaccharide and D-galactosamine-induced liver injury. Cell Tissue Res 2015; 363:371-83. [PMID: 26267221 DOI: 10.1007/s00441-015-2252-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
Tumor necrosis factor (TNF)-α exists in two bioactive forms, a 26-kDa transmembrane form (tmTNF-α) and a 17-kDa soluble form (sTNF-α). sTNF-α has been recognized as a key regulator of hepatitis; however, serum sTNF-α disappears in mice during the development of severe liver injury, and high levels of serum sTNF-α do not necessarily result in liver damage. Interestingly, in a mouse model of acute hepatitis, we have found that tmTNF-α expression on Kupffer cells (KCs) significantly increases when mice develop severe liver injury caused by lipopolysaccharide (LPS)/D-galactosamine (D-gal), and the level of tmTNF-α expression is positively related to the activity of serum transaminases. Therefore, we hypothesized that KC-expressed tmTNF-α constitutes a pathomechanism in hepatitis and have explored the role of tmTNF-α in this disease model. Here, we have compared the impact of KCs(tmTNFlow) and KCs(tmTNFhigh) on acute hepatitis in vivo and ex vivo and have further demonstrated that KCs(tmTNFhigh), rather than KCs(tmTNFlow), not only exhibit an imbalance in secretion of pro- and anti-inflammatory cytokines, favoring inflammatory response and exacerbating liver injury, but also induce hepatocellular apoptosis via tmTNF-α and the expression of another pro-apoptotic factor, Fas ligand. Our data suggest that KC(tmTNFhigh) is a major contributor to liver injury in LPS/D-gal-induced hepatitis.
Collapse
Affiliation(s)
- Peng Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Wenjing Zhou
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Chenxi Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Meng Zhang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Yaping Jiang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Rui Jiang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Hongping Ba
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Cheng Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Jing Wang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Bingjiao Yin
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Feili Gong
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China
| | - Zhuoya Li
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan, Hubei, 430030, People's Republic of China.
| |
Collapse
|
44
|
Chen W, Han C, Zhang J, Song K, Wang Y, Wu T. miR-150 Deficiency Protects against FAS-Induced Acute Liver Injury in Mice through Regulation of AKT. PLoS One 2015. [PMID: 26196694 PMCID: PMC4510058 DOI: 10.1371/journal.pone.0132734] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Although miR-150 is implicated in the regulation of immune cell differentiation and activation, it remains unknown whether miR-150 is involved in liver biology and disease. This study was performed to explore the potential role of miR-150 in LPS/D-GalN and Fas-induced liver injuries by using wild type and miR-150 knockout (KO) mice. Whereas knockout of miR-150 did not significantly alter LPS/D-GalN-induced animal death and liver injury, it protected against Fas-induced liver injury and mortality. The Jo2-induced increase in serum transaminases, apoptotic hepatocytes, PARP cleavage, as well as caspase-3/7, caspase-8, and caspase-9 activities were significantly attenuated in miR-150 KO mice. The liver tissues from Jo2-treated miR-150 KO mice expressed higher levels of Akt1, Akt2, total Akt, as well as p-Akt(Ser473) compared to the wild type livers. Pretreatment with the Akt inhibitor V reversed Jo2-induced liver injury in miR-150 KO mice. The primary hepatocytes isolated from miR-150 KO mice also showed protection against Fas-induced apoptosis in vitro (characterized by less prominent PARP cleavage, less nuclear fragmentation and less caspase activation) in comparison to hepatocytes from wild type mice. Luciferase reporter assays in hepatocytes transfected with the Akt1 or Akt2 3’-UTR reporter constructs (with or without mutation of miR-150 binding site) established Akt1 and Akt2 as direct targets of miR-150. Tail vein injection of lentiviral particles containing pre-miR-150 enhanced Jo2-induced liver injury in miR-150 KO mice. These findings demonstrate that miR-150 deficiency prevents Fas-induced hepatocyte apoptosis and liver injury through regulation of the Akt pathway.
Collapse
Affiliation(s)
- Weina Chen
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
| | - Chang Han
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
| | - Jinqiang Zhang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
| | - Kyoungsub Song
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
- Department of Gastroenterology and Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine,1430 Tulane Avenue SL-79, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
45
|
Sharifov OF, Xu X, Gaggar A, Tabengwa EM, White CR, Palgunachari MN, Anantharamaiah GM, Gupta H. L-4F inhibits lipopolysaccharide-mediated activation of primary human neutrophils. Inflammation 2015; 37:1401-12. [PMID: 24647607 DOI: 10.1007/s10753-014-9864-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Human apolipoprotein A-I (apoA-I) mimetic L-4F inhibits acute inflammation in endotoxemic animals. Since neutrophils play a crucial role in septic inflammation, we examined the effects of L-4F, compared to apoA-I, on lipopolysaccharide (LPS)-mediated activation of human neutrophils. We performed bioassays in human blood, isolated human neutrophils (incubated in 50 % donor plasma), and isolated human leukocytes (incubated in 5 and 50 % plasma) in vitro. In whole blood, both L-4F and apoA-I inhibited LPS-mediated elevation of TNF-α and IL-6. In LPS-stimulated neutrophils, L-4F and apoA-I (40 μg/ml) also decreased myeloperoxidase and TNF-α levels; however, L-4F tended to be superior in inhibiting LPS-mediated increase in IL-6 levels, membrane lipid rafts abundance and CD11b expression. In parallel experiments, when TNF-α and IL-8, instead of LPS, was used for cell stimulation, L-4F and/or apoA-I revealed only limited efficacy. In LPS-stimulated leukocytes, L-4F was as effective as apoA-I in reducing superoxide formation in 50 % donor plasma, and more effective in 5 % donor plasma (P<0.05). Limulus ambocyte lysate (LAL) and surface plasmon resonance assays showed that L-4F neutralizes LAL endotoxin activity more effectively than apoA-I (P<0.05) likely due to avid binding to LPS. We conclude that (1) direct binding/neutralization of LPS is a major mechanism of L-4F in vitro; (2) while L-4F has similar efficacy to apoA-I in anti-endotoxin effects in whole blood, it demonstrates superior efficacy to apoA-I in aqueous solutions and fluids with limited plasma components. This study rationalizes the utility of L-4F in the treatment of inflammation that is mediated by endotoxin-activated neutrophils.
Collapse
Affiliation(s)
- Oleg F Sharifov
- Department of Medicine, University of Alabama at Birmingham, BDB-101, 1808 7th Avenue South, Birmingham, AL, 35294-0012, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Lactobacillus fermentum ZYL0401 Attenuates Lipopolysaccharide-Induced Hepatic TNF-α Expression and Liver Injury via an IL-10- and PGE2-EP4-Dependent Mechanism. PLoS One 2015; 10:e0126520. [PMID: 25978374 PMCID: PMC4433256 DOI: 10.1371/journal.pone.0126520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 04/03/2015] [Indexed: 02/07/2023] Open
Abstract
Lipopolysaccharide (LPS) has essential role in the pathogenesis of D-galactosamine-sensitized animal models and alcoholic liver diseases of humans, by stimulating release of pro-inflammatory mediators that cause hepatic damage and intestinal barrier impairment. Oral pretreatment of probiotics has been shown to attenuate LPS-induced hepatic injury, but it is unclear whether the effect is direct or due to improvement in the intestinal barrier. The present study tested the hypothesis that pretreatment with probiotics enables the liver to withstand directly LPS-induced hepatic injury and inflammation. In a mouse model of LPS-induced hepatic injury, the levels of hepatic tumor necrosis factor-alpha (TNF-α) and serum alanine aminotransferase (ALT) of mice with depleted intestinal commensal bacteria were not significantly different from that of the control models. Pre-feeding mice for 10 days with Lactobacillus fermentum ZYL0401 (LF41), significantly alleviated LPS-induced hepatic TNF-α expression and liver damage. After LF41 pretreatment, mice had dramatically more L.fermentum-specific DNA in the ileum, significantly higher levels of ileal cyclooxygenase (COX)-2 and interleukin 10 (IL-10) and hepatic prostaglandin E2 (PGE2). However, hepatic COX-1, COX-2, and IL-10 protein levels were not changed after the pretreatment. There were also higher hepatic IL-10 protein levels after LPS challenge in LF41-pretreaed mice than in the control mice. Attenuation of hepatic TNF-α was mediated via the PGE2/E prostanoid 4 (EP4) pathway, and serum ALT levels were attenuated in an IL-10-dependent manner. A COX-2 blockade abolished the increase in hepatic PGE2 and IL-10 associated with LF41. In LF41-pretreated mice, a blockade of IL-10 caused COX-2-dependent promotion of hepatic PGE2, without affecting hepatic COX-2levels. In LF41-pretreated mice, COX2 prevented enhancing TNF-α expression in both hepatic mononuclear cells and the ileum, and averted TNF-α-mediated increase in intestinal permeability. Together, we demonstrated that LF41 pre-feeding enabled the liver to alleviate LPS-induced hepatic TNF-α expression and injury via a PGE2-EP4- and IL-10-dependent mechanism.
Collapse
|
47
|
Maegawa Y, Mochizuki S, Miyamoto N, Sanada Y, Sakurai K. Application toward Drug Delivery System Using ^|^beta;-1,3-Glucan. TRENDS GLYCOSCI GLYC 2015. [DOI: 10.4052/tigg.27.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
48
|
Kim SJ, Cho HI, Kim SJ, Kim JS, Kwak JH, Lee DU, Lee SK, Lee SM. Protective effects of lupeol against D-galactosamine and lipopolysaccharide-induced fulminant hepatic failure in mice. JOURNAL OF NATURAL PRODUCTS 2014; 77:2383-2388. [PMID: 25325613 DOI: 10.1021/np500296b] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
This study examined the hepatoprotective effects of lupeol (1, a major active triterpenoid isolated from Adenophora triphylla var. japonica) against d-galactosamine (GalN) and lipopolysaccharide (LPS)-induced fulminant hepatic failure. Mice were orally administered 1 (25, 50, and 100 mg/kg; dissolved in olive oil) 1 h before GalN (800 mg/kg)/LPS (40 μg/kg) treatment. Treatment with GalN/LPS resulted in increased levels of serum alanine aminotransferase, tumor necrosis factor (TNF)-α, and interleukin (IL)-6, as well as increased mortality, all of which were attenuated by treatment with 1. In addition, levels of toll-like receptor (TLR)4, myeloid differentiation primary response gene 88, TIR-domain-containing adapter-inducing interferon-β (TRIF), IL-1 receptor-associated kinase (IRAK)-1, and TNF receptor associated factor 6 protein expression were increased by GalN/LPS. These increases, except TRIF, were attenuated by 1. Interestingly, 1 augmented GalN/LPS-mediated increases in the protein expression of IRAK-M, a negative regulator of TLR signaling. Following GalN/LPS treatment, nuclear translocation of nuclear factor-κB and the levels of TNF-α and IL-6 mRNA expression increased, which were attenuated by 1. Together, the present findings suggest that lupeol (1) ameliorates GalN/LPS-induced liver injury, which may be due to inhibition of IRAK-mediated TLR inflammatory signaling.
Collapse
Affiliation(s)
- So-Jin Kim
- School of Pharmacy, Sungkyunkwan University , Suwon, 440-746, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Necrostatin-1 protects against reactive oxygen species (ROS)-induced hepatotoxicity in acetaminophen-induced acute liver failure. FEBS Open Bio 2014; 4:777-87. [PMID: 25349782 PMCID: PMC4208088 DOI: 10.1016/j.fob.2014.08.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 08/29/2014] [Accepted: 08/30/2014] [Indexed: 12/13/2022] Open
Abstract
RIPK-dependent necrosis is involved in acetaminophen (APAP)-induced hepatotoxicity. Necrostatin-1 (Nec-1) protects mice against APAP-induced acute liver damage. Nec-1 suppresses APAP-induced ROS generation in hepatocytes. Nec-1 promotes resistance to oxidative stress in hepatocytes.
Excessive acetaminophen (APAP) use is one of the most common causes of acute liver failure. Various types of cell death in the damaged liver are linked to APAP-induced hepatotoxicity, and, of these, necrotic cell death of hepatocytes has been shown to be involved in disease pathogenesis. Until recently, necrosis was commonly considered to be a random and unregulated form of cell death; however, recent studies have identified a previously unknown form of programmed necrosis called receptor-interacting protein kinase (RIPK)-dependent necrosis (or necroptosis), which is controlled by the kinases RIPK1 and RIPK3. Although RIPK-dependent necrosis has been implicated in a variety of disease states, including atherosclerosis, myocardial organ damage, stroke, ischemia–reperfusion injury, pancreatitis, and inflammatory bowel disease. However its involvement in APAP-induced hepatocyte necrosis remains elusive. Here, we showed that RIPK1 phosphorylation, which is a hallmark of RIPK-dependent necrosis, was induced by APAP, and the expression pattern of RIPK1 and RIPK3 in the liver overlapped with that of CYP2E1, whose activity around the central vein area has been demonstrated to be critical for the development of APAP-induced hepatic injury. Moreover, a RIPK1 inhibitor ameliorated APAP-induced hepatotoxicity in an animal model, which was underscored by significant suppression of the release of hepatic enzymes and cytokine expression levels. RIPK1 inhibition decreased reactive oxygen species levels produced in APAP-injured hepatocytes, whereas CYP2E1 expression and the depletion rate of total glutathione were unaffected. Of note, RIPK1 inhibition also conferred resistance to oxidative stress in hepatocytes. These data collectively demonstrated a RIPK-dependent necrotic mechanism operates in the APAP-injured liver and inhibition of this pathway may be beneficial for APAP-induced fulminant hepatic failure.
Collapse
Key Words
- ABTS, 2,2′-azino-bis (3-ethylbenzothiazoline)-6-sulfonic acid
- ALF, acute liver failure
- ALT, alanine aminotransferase
- APAP, acetaminophen
- AST, aspartate aminotransferase
- Acetaminophen
- Acute liver failure
- CM-H2DCFDA, 5-(and-6)-chloromethyl-2′,7′-dichlorodihydrofluorescein diacetate, acetyl ester
- CXCL1, chemokine (C-X-C motif) ligand 1
- CYP2E1, cytochrome P450 2E1
- DMSO, dimethyl sulfoxide
- Drp1, dynamin-related protein 1
- FBS, fetal bovine serum
- GSH, glutathione
- Hepatocytes
- LDH, lactate dehydrogenase
- NAPQI, N-acetyl-p-benzoquinone
- NO, nitric oxide
- Nec-1, necrostatin-1
- Necroptosis
- PGAM5, phosphoglycerate mutase family member 5
- PI, propidium iodide
- RIPK, receptor-interacting protein kinase
- RIPK-dependent necrosis
- ROS, reactive oxygen species
- Reactive oxygen species
- SNAP, S-nitroso-N-acetyl-dl-penicillamine
- WST-8, 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium
- bFGF, basic fibroblast growth factor
- λPP, lambda protein phosphatase
Collapse
|
50
|
Zhang J, Xu L, Zhang L, Ying Z, Su W, Wang T. Curcumin attenuates D-galactosamine/lipopolysaccharide-induced liver injury and mitochondrial dysfunction in mice. J Nutr 2014; 144:1211-8. [PMID: 24899159 DOI: 10.3945/jn.114.193573] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Curcumin, a naturally occurring antioxidant, has various beneficial effects in the treatment of human diseases. However, little information regarding the protection it provides against acute liver injury is available. The present study investigated the protective effects of curcumin against D-galactosamine (D-GalN)/lipopolysaccharide (LPS)-induced acute liver injury in mice. A total of 40 male Kunming mice were randomly assigned to 5 groups: 1) mice administered saline vehicle injection (control), 2) mice administered 200 mg/kg body weight (BW) curcumin by i.p. injection (CUR), 3) mice administered D-GalN/LPS (700 mg and 5 μg/kg BW) via i.p. injection (GL), 4) mice administered 200 mg/kg BW curcumin i.p. 1 h before D-GalN/LPS injection (CUR-GL), and 5) mice administered 200 mg/kg BW curcumin i.p. 1 h after D-GalN/LPS injection (GL-CUR). Twenty h after D-GalN/LPS injection, serum alanine aminotransferase activities were 18.5% and 13.5% lower (P < 0.05) and aspartate aminotransferase (AST) activities were 26.6% and 9.6% lower (P < 0.05) in the CUR-GL and GL-CUR groups, respectively, than in the GL group. The CUR-GL and GL-CUR groups had 64.4% and 15.0% higher (P < 0.05) mitochondrial membrane potentials, respectively, and the CUR-GL group had a 44.7% lower reactive oxygen species concentration than the GL group (P < 0.05). Mitochondrial manganese superoxide dismutase activities were 111% and 77.9% higher (P < 0.05) and the percentages of necrotic cells were 47.0% and 32.4% lower (P < 0.05) in the CUR-GL and GL-CUR groups, respectively, than in the GL group. Liver mRNA levels of sirtuin 1 (Sirt1) were 56.4% lower (P < 0.05) in the CUR-GL group than in the GL group. Moreover, compared with the GL-CUR group, the CUR-GL group had an 18.7% lower serum AST activity, a 31.7% lower mitochondrial malondialdehyde concentration, a 36.0% lower hepatic reactive oxygen species concentration, and a 43.0% higher mitochondrial membrane potential. These results suggested that curcumin protects against D-GalN/LPS-induced liver damage by the enhancing antioxidant defense system, attenuating mitochondrial dysfunction and inhibiting apoptosis. This was especially true for curcumin pretreatment, which highlighted its promise as a preventive treatment for acute liver injury in clinical settings.
Collapse
Affiliation(s)
- Jingfei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Li Xu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Zhixiong Ying
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Weipeng Su
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, People's Republic of China
| |
Collapse
|