1
|
Acuña-Pilarte K, Reichert EC, Green YS, Halberg LMT, Golkowski M, Maguire KM, Mimche PN, Kamdem SD, Hu PA, Wright J, Ducker GS, Voth WP, O'Connell RM, McFarland SA, Egal ESA, Chaix A, Summers SA, Reelitz JW, Maschek JA, Cox JE, Evason KJ, Koh MY. HAF prevents hepatocyte apoptosis and progression to MASH and HCC through transcriptional regulation of the NF-κB pathway. Hepatology 2024:01515467-990000000-01023. [PMID: 39255518 DOI: 10.1097/hep.0000000000001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/30/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND AND AIMS HCC incidence is increasing worldwide due to the obesity epidemic, which drives metabolic dysfunction-associated steatohepatitis (MASH) that can lead to HCC. However, the molecular pathways driving MASH-HCC are poorly understood. We have previously reported that male mice with haploinsufficiency of hypoxia-associated factor (HAF) ( SART1+/ - ) spontaneously develop MASH-HCC. However, the cell type(s) responsible for HCC associated with HAF loss are unclear. APPROACH AND RESULTS We generated SART1 -floxed mice, which were crossed with mice expressing Cre recombinase within hepatocytes (Alb-Cre; hepS -/- ) or myeloid cells (LysM-Cre, macS -/- ). HepS - / - mice (both male and female) developed HCC associated with profound inflammatory and lipid dysregulation, suggesting that HAF protects against HCC primarily within hepatocytes. HAF-deficient hepatocytes showed decreased P-p65 and P-p50 in many components of the NF-κB pathway, which was recapitulated using HAF small interfering RNA in vitro. HAF depletion also triggered apoptosis, suggesting that HAF protects against HCC by suppressing hepatocyte apoptosis. We show that HAF regulates NF-κB activity by regulating the transcription of TRADD and RIPK1 . Mice fed a high-fat diet showed marked suppression of HAF, P-p65, and TRADD within their livers after 26 weeks but showed profound upregulation of these proteins after 40 weeks, implicating deregulation of the HAF-NF-κB axis in the progression to MASH. In humans, HAF was significantly decreased in livers with simple steatosis but significantly increased in HCC compared with normal liver. CONCLUSIONS HAF is a novel transcriptional regulator of the NF-κB pathway and is a key determinant of cell fate during progression to MASH and MASH-HCC.
Collapse
Affiliation(s)
- Karen Acuña-Pilarte
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Ethan C Reichert
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Yangsook Song Green
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Lily M-T Halberg
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Martin Golkowski
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | | | - Patrice N Mimche
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | | | - Po-An Hu
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Jillian Wright
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Gregory S Ducker
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Warren P Voth
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Ryan M O'Connell
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Sydney A McFarland
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| | - Erika Said Abu Egal
- Biorepository and Molecular Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Amandine Chaix
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah, USA
| | - Jordan W Reelitz
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - John Alan Maschek
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - James E Cox
- Department of Biochemistry, University of Utah, Salt Lake City, Utah, USA
| | - Kimberley J Evason
- Department of Anatomic Pathology, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Mei Yee Koh
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah, USA
| |
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
|
Pilarte KA, Reichert EC, Green YS, Halberg LMT, McFarland SA, Mimche PN, Golkowski M, Kamdem SD, Maguire KM, Summers SA, Maschek JA, Reelitz JW, Cox JE, Evason KJ, Koh MY. HAF Prevents Hepatocyte Apoptosis and Hepatocellular Carcinoma through Transcriptional Regulation of the NF-κB pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574894. [PMID: 38260413 PMCID: PMC10802431 DOI: 10.1101/2024.01.09.574894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Background Hepatocellular carcinoma (HCC) incidence is increasing worldwide due to the obesity epidemic, which drives metabolic dysfunction-associated steatohepatitis (MASH) that can lead to HCC. However, the molecular pathways that lead to MASH-HCC are poorly understood. We have previously reported that male mice with global haploinsufficiency of hypoxia-associated factor, HAF ( SART1 +/ - ) spontaneously develop MASH/HCC. However, the cell type(s) responsible for HCC associated with HAF loss are unclear. Results SART1 -floxed mice were crossed with mice expressing Cre-recombinase within hepatocytes (Alb-Cre; hepS -/- ) or macrophages (LysM-Cre, macS -/- ). Only hepS -/- mice (both male and female) developed HCC suggesting that HAF protects against HCC primarily within hepatocytes. HAF-deficient macrophages showed decreased P-p65 and P-p50 and in many major components of the NF-κB pathway, which was recapitulated using HAF siRNA in vitro . HAF depletion increased apoptosis both in vitro and in vivo , suggesting that HAF mediates a tumor suppressor role by suppressing hepatocyte apoptosis. We show that HAF regulates NF-κB activity by controlling transcription of TRADD and RIPK1 . Mice fed a high-fat diet (HFD) showed marked suppression of HAF, P-p65 and TRADD within their livers after 26 weeks, but manifest profound upregulation of HAF, P-65 and TRADD within their livers after 40 weeks of HFD, implicating deregulation of the HAF-NF-κB axis in the progression to MASH. In humans, HAF was significantly decreased in livers with simple steatosis but significantly increased in HCC compared to normal liver. Conclusions HAF is novel transcriptional regulator of the NF-κB pathway that protects against hepatocyte apoptosis and is a key determinant of cell fate during progression to MASH and MASH-HCC.
Collapse
|
4
|
Siegmund D, Zaitseva O, Wajant H. Fn14 and TNFR2 as regulators of cytotoxic TNFR1 signaling. Front Cell Dev Biol 2023; 11:1267837. [PMID: 38020877 PMCID: PMC10657838 DOI: 10.3389/fcell.2023.1267837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Tumor necrosis factor (TNF) receptor 1 (TNFR1), TNFR2 and fibroblast growth factor-inducible 14 (Fn14) belong to the TNF receptor superfamily (TNFRSF). From a structural point of view, TNFR1 is a prototypic death domain (DD)-containing receptor. In contrast to other prominent death receptors, such as CD95/Fas and the two TRAIL death receptors DR4 and DR5, however, liganded TNFR1 does not instruct the formation of a plasma membrane-associated death inducing signaling complex converting procaspase-8 into highly active mature heterotetrameric caspase-8 molecules. Instead, liganded TNFR1 recruits the DD-containing cytoplasmic signaling proteins TRADD and RIPK1 and empowers these proteins to trigger cell death signaling by cytosolic complexes after their release from the TNFR1 signaling complex. The activity and quality (apoptosis versus necroptosis) of TNF-induced cell death signaling is controlled by caspase-8, the caspase-8 regulatory FLIP proteins, TRAF2, RIPK1 and the RIPK1-ubiquitinating E3 ligases cIAP1 and cIAP2. TNFR2 and Fn14 efficiently recruit TRAF2 along with the TRAF2 binding partners cIAP1 and cIAP2 and can thereby limit the availability of these molecules for other TRAF2/cIAP1/2-utilizing proteins including TNFR1. Accordingly, at the cellular level engagement of TNFR2 or Fn14 inhibits TNFR1-induced RIPK1-mediated effects reaching from activation of the classical NFκB pathway to induction of apoptosis and necroptosis. In this review, we summarize the effects of TNFR2- and Fn14-mediated depletion of TRAF2 and the cIAP1/2 on TNFR1 signaling at the molecular level and discuss the consequences this has in vivo.
Collapse
Affiliation(s)
| | | | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Huyghe J, Priem D, Bertrand MJM. Cell death checkpoints in the TNF pathway. Trends Immunol 2023:S1471-4906(23)00105-9. [PMID: 37357102 DOI: 10.1016/j.it.2023.05.007] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/27/2023]
Abstract
Tumor necrosis factor (TNF) plays a central role in orchestrating mammalian inflammatory responses. It promotes inflammation either directly by inducing inflammatory gene expression or indirectly by triggering cell death. TNF-mediated cell death-driven inflammation can be beneficial during infection by providing cell-extrinsic signals that help to mount proper immune responses. Uncontrolled cell death caused by TNF is instead highly detrimental and is believed to cause several human autoimmune diseases. Death is not the default response to TNF sensing. Molecular brakes, or cell death checkpoints, actively repress TNF cytotoxicity to protect the organism from its detrimental consequences. These checkpoints therefore constitute essential safeguards against inflammatory diseases. Recent advances in the field have revealed the existence of several new and unexpected brakes against TNF cytotoxicity and pathogenicity.
Collapse
Affiliation(s)
- Jon Huyghe
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Dario Priem
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Mathieu J M Bertrand
- Cell Death and Inflammation Unit, Vlaams Instituut voor Biotechnologie (VIB) Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
6
|
Liu P, Li Y, Wang W, Bai Y, Jia H, Yuan Z, Yang Z. Role and mechanisms of the NF-ĸB signaling pathway in various developmental processes. Biomed Pharmacother 2022; 153:113513. [DOI: 10.1016/j.biopha.2022.113513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 11/02/2022] Open
|
7
|
Hövelmeyer N, Schmidt-Supprian M, Ohnmacht C. NF-κB in control of regulatory T cell development, identity, and function. J Mol Med (Berl) 2022; 100:985-995. [PMID: 35672519 PMCID: PMC9213371 DOI: 10.1007/s00109-022-02215-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 12/11/2022]
Abstract
Regulatory T cells (Treg cells) act as a major rheostat regulating the strength of immune responses, enabling tolerance of harmless foreign antigens, and preventing the development of pathogenic immune responses in various disease settings such as cancer and autoimmunity. Treg cells are present in all lymphoid and non-lymphoid tissues, and the latter often fulfill important tasks required for the physiology of their host organ. The activation of NF-κB transcription factors is a central pathway for the reprogramming of gene expression in response to inflammatory but also homeostatic cues. Genetic mouse models have revealed essential functions for NF-κB transcription factors in modulating Treg development and function, with some of these mechanistic insights confirmed by recent studies analyzing Treg cells from patients harboring point mutations in the genes encoding NF-κB proteins. Molecular insights into the NF-κB pathway in Treg cells hold substantial promise for novel therapeutic strategies to manipulate dysfunctional or inadequate cell numbers of immunosuppressive Treg cells in autoimmunity or cancer. Here, we provide an overview of the manifold roles that NF-κB factors exert in Treg cells.
Collapse
Affiliation(s)
- Nadine Hövelmeyer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
- Germany Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.
| | - Marc Schmidt-Supprian
- Institute for Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University Munich, Munich, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| | - Caspar Ohnmacht
- Center for Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany.
| |
Collapse
|
8
|
Linoleic acid reduces apoptosis via NF-κB during the in vitro development of induced parthenogenic porcine embryos. Theriogenology 2022; 187:173-181. [PMID: 35596974 DOI: 10.1016/j.theriogenology.2022.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022]
Abstract
Fatty acid has a various role in preimplantation embryo development. Especially, Linoleic acid, polyunsaturated fatty acid, has been reported to affect the apoptosis pathway via nuclear transcription factor-kappa B. But to date, the function of NF-κB has not been demonstrated in porcine preimplantation embryos. We demonstrated that linoleic acid had a positive effect on embryo development at a certain concentration(25 μM), but developmental failure was observed at higher concentration. Furthermore, the expression level of NF-κB increased, unlike that of IL-6, as the concentration of linoleic acid increased. Interestingly, the concentration of NF-κB was found to increase even at the concentration of linoleic acid at which embryo development decreased. We found that pro-apoptotic gene expression was downregulated in the linoleic acid-treated group. It was also found that MCL-1, an anti-apoptotic gene known to be unaffected by IL-6, was found to be increased at the mRNA level in the linoleic acid-treated group. As the concentration of NF-kB increased, the nuclear translocation of C-JUN gradually increased dependent on the linoleic acid concentration. It was confirmed that NF-κB is an important factor in porcine embryos by treated ammonium pyrrolidinedithiocarbamate (APDC 0.1 μM, an inhibitor of NF-κB) affected NF-κB protein expression, IL-6 expression, and blastocyst production. These data supported porcine embryos can use exogenous linoleic acid as a metabolic energy source via NF-κB.
Collapse
|
9
|
Lecerf K, Koboldt DC, Kuehn HS, Jayaraman V, Lee K, Mihalic Mosher T, Yonkof JR, Mori M, Hickey SE, Franklin S, Drew J, Akoghlanian S, Sivaraman V, Rosenzweig SD, Wilson RK, Abraham RS. Case report and review of the literature: immune dysregulation in a large familial cohort due to a novel pathogenic RELA variant. Rheumatology (Oxford) 2022; 62:347-359. [PMID: 35412596 PMCID: PMC9960492 DOI: 10.1093/rheumatology/keac227] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/23/2022] [Indexed: 12/27/2022] Open
Abstract
OBJECTIVE To explore and define the molecular cause(s) of a multi-generational kindred affected by Bechet's-like mucocutaneous ulcerations and immune dysregulation. METHODS Whole genome sequencing and confirmatory Sanger sequencing were performed. Components of the NFκB pathway were quantified by immunoblotting, and function was assessed by cytokine production (IL-6, TNF-α, IL-1β) after lipopolysaccharide (LPS) stimulation. Detailed immunophenotyping of T-cell and B-cell subsets was performed in four patients from this cohort. RESULTS A novel variant in the RELA gene, p. Tyr349LeufsTer13, was identified. This variant results in premature truncation of the protein before the serine (S) 536 residue, a key phosphorylation site, resulting in enhanced degradation of the p65 protein. Immunoblotting revealed significantly decreased phosphorylated [p]p65 and pIκBα. The decrease in [p]p65 may suggest reduced heterodimer formation between p50/p65 (NFκB1/RelA). Immunophenotyping revealed decreased naïve T cells, increased memory T cells, and expanded senescent T-cell populations in one patient (P1). P1 also had substantially higher IL-6 and TNF-α levels post-stimulation compared with the other three patients. CONCLUSION Family members with this novel RELA variant have a clinical phenotype similar to other reported RELA cases with predominant chronic mucocutaneous ulceration; however, the clinical phenotype broadens to include Behçet's syndrome and IBD. Here we describe the clinical, immunological and genetic evaluation of a large kindred to further expand identification of patients with autosomal dominant RELA deficiency, facilitating earlier diagnosis and intervention. The functional impairment of the canonical NFκB pathway suggests that this variant is causal for the clinical phenotype in these patients.
Collapse
Affiliation(s)
- Kelsey Lecerf
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital,Division of Allergy and Immunology, Department of Otolaryngology, The Ohio State University Wexner Medical Center
| | - Daniel C Koboldt
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Hye Sun Kuehn
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD
| | - Vijayakumar Jayaraman
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Kristy Lee
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH,Department of Pathology, The Ohio State University Wexner College of Medicine, Columbus, OH
| | - Theresa Mihalic Mosher
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH,Ambry Genetics, Aliso Viejo, CA
| | | | - Mari Mori
- Division of Genetic and Genomic Medicine
| | | | - Samuel Franklin
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Joanne Drew
- Division of Pediatric Rheumatology, Department of Pediatrics
| | | | - Vidya Sivaraman
- Division of Pediatric Rheumatology, Department of Pediatrics
| | - Sergio D Rosenzweig
- Immunology Service, Department of Laboratory Medicine, NIH Clinical Center, Bethesda, MD
| | - Richard K Wilson
- The Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH
| | - Roshini S Abraham
- Correspondence to: Roshini S. Abraham, Nationwide Children’s Hospital, 700 Children’s Drive, Columbus, OH-43205, USA. E-mail:
| |
Collapse
|
10
|
IKKα plays a major role in canonical NF-kB signalling in colorectal cells. Biochem J 2022; 479:305-325. [PMID: 35029639 PMCID: PMC8883499 DOI: 10.1042/bcj20210783] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 11/17/2022]
Abstract
Inhibitor of kappa B (IκB) kinase β (IKKβ) has long been viewed as the dominant IKK in the canonical nuclear factor-κB (NF-κB) signalling pathway, with IKKα being more important in non-canonical NF-κB activation. Here we have investigated the role of IKKα and IKKβ in canonical NF-κB activation in colorectal cells using CRISPR–Cas9 knock-out cell lines, siRNA and selective IKKβ inhibitors. IKKα and IKKβ were redundant for IκBα phosphorylation and turnover since loss of IKKα or IKKβ alone had little (SW620 cells) or no (HCT116 cells) effect. However, in HCT116 cells IKKα was the dominant IKK required for basal phosphorylation of p65 at S536, stimulated phosphorylation of p65 at S468, nuclear translocation of p65 and the NF-κB-dependent transcriptional response to both TNFα and IL-1α. In these cells, IKKβ was far less efficient at compensating for the loss of IKKα than IKKα was able to compensate for the loss of IKKβ. This was confirmed when siRNA was used to knock-down the non-targeted kinase in single KO cells. Critically, the selective IKKβ inhibitor BIX02514 confirmed these observations in WT cells and similar results were seen in SW620 cells. Notably, whilst IKKα loss strongly inhibited TNFα-dependent p65 nuclear translocation, IKKα and IKKβ contributed equally to c-Rel nuclear translocation indicating that different NF-κB subunits exhibit different dependencies on these IKKs. These results demonstrate a major role for IKKα in canonical NF-κB signalling in colorectal cells and may be relevant to efforts to design IKK inhibitors, which have focused largely on IKKβ to date.
Collapse
|
11
|
Li X, Hu Y. Attribution of NF-κB Activity to CHUK/IKKα-Involved Carcinogenesis. Cancers (Basel) 2021; 13:cancers13061411. [PMID: 33808757 PMCID: PMC8003426 DOI: 10.3390/cancers13061411] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary CHUK/IKKα has emerged as a novel tumor suppressor in several organs of humans and mice. In general, activation of NF-κB promotes inflammation and tumorigenesis. IKKα reduction stimulates inflammatory responses including NF-κB’s targets and NF-κB-independent pathways for tumor promotion. Specific phenomena from genetically-modified mice and human TCGA database show the crosstalk between IKKα and NF-κB although their nature paths for normal organ development and the disease and cancer pathogenesis remains largely under investigation. In this review, we focus on the interplay between IKKα and NF-κB signaling during carcinogenesis. A better understanding of their relationship will provide insight into therapeutic targets of cancer. Abstract Studies analyzing human cancer genome sequences and genetically modified mouse models have extensively expanded our understanding of human tumorigenesis, even challenging or reversing the dogma of certain genes as originally characterized by in vitro studies. Inhibitor-κB kinase α (IKKα), which is encoded by the conserved helix-loop-helix ubiquitous kinase (CHUK) gene, is first identified as a serine/threonine protein kinase in the inhibitor-κB kinase complex (IKK), which is composed of IKKα, IKKβ, and IKKγ (NEMO). IKK phosphorylates serine residues 32 and 36 of IκBα, a nuclear factor-κB (NF-κB) inhibitor, to induce IκBα protein degradation, resulting in the nuclear translocation of NF-κB dimers that function as transcriptional factors to regulate immunity, infection, lymphoid organ/cell development, cell death/growth, and tumorigenesis. NF-κB and IKK are broadly and differentially expressed in the cells of our body. For a long time, the idea that the IKK complex acts as a direct upstream activator of NF-κB in carcinogenesis has been predominately accepted in the field. Surprisingly, IKKα has emerged as a novel suppressor for skin, lung, esophageal, and nasopharyngeal squamous cell carcinoma, as well as lung and pancreatic adenocarcinoma (ADC). Thus, Ikkα loss is a tumor driver in mice. On the other hand, lacking the RANKL/RANK/IKKα pathway impairs mammary gland development and attenuates oncogene- and chemical carcinogen-induced breast and prostate tumorigenesis and metastasis. In general, NF-κB activation leads one of the major inflammatory pathways and stimulates tumorigenesis. Since IKKα and NF-κB play significant roles in human health, revealing the interplay between them greatly benefits the diagnosis, treatment, and prevention of human cancer. In this review, we discuss the intriguing attribution of NF-κB to CHUK/IKKα-involved carcinogenesis.
Collapse
|
12
|
Ding J, Zhao J, Huan L, Liu Y, Qiao Y, Wang Z, Chen Z, Huang S, Zhao Y, He X. Inflammation-Induced Long Intergenic Noncoding RNA (LINC00665) Increases Malignancy Through Activating the Double-Stranded RNA-Activated Protein Kinase/Nuclear Factor Kappa B Pathway in Hepatocellular Carcinoma. Hepatology 2020; 72:1666-1681. [PMID: 32083756 DOI: 10.1002/hep.31195] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 01/09/2020] [Accepted: 01/26/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIMS The nuclear factor kappa B (NF-κB) signaling pathway is important for linking inflammation and tumorigenesis. Here, we characterized an NF-κB signaling activation-induced long intergenic noncoding (LINC) RNA in hepatocellular carcinoma (HCC), LINC00665, that contributes to the enhanced cell proliferation of HCC cells both in vitro and in vivo. APPROACH AND RESULTS LINC00665 physically interacts with the double-stranded RNA (dsRNA)-activated protein kinase (PKR), enhances its activation, and maintains its protein stability by blocking ubiquitin/proteasome-dependent degradation, resulting in a positive feedback regulation of NF-κB signaling in HCC cells. Notably, patients with HCC and higher LINC00665 have poorer outcomes in the clinic. CONCLUSIONS Our findings indicate that LINC00665 is involved in the NF-κB signaling activation in HCC cells and that the inflammatory LINC00665/PKR/NF-κB loop plays important oncogenic roles in hepatic cancer progression and may be a potential therapeutic target.
Collapse
Affiliation(s)
- Jie Ding
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingjing Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lin Huan
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yizhe Liu
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yejun Qiao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhen Wang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiao Chen
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shenglin Huang
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Yingjun Zhao
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Xianghuo He
- Fudan University Shanghai Cancer Center and Institutes of Biomedical Sciences, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Comprehensive Targeted Sequencing Identifies Monogenic Disorders in Patients With Early-onset Refractory Diarrhea. J Pediatr Gastroenterol Nutr 2020; 71:333-339. [PMID: 32487952 DOI: 10.1097/mpg.0000000000002796] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Causes of early-onset refractory diarrhea include exudative diarrhea associated with very early-onset inflammatory bowel diseases, osmotic or secretory diarrhea, and protein-losing enteropathy. Monogenic disorders are included in these diseases, yet a comprehensive genetic analysis has not been fully established. METHODS We established targeted gene panels covering all responsible genes for early-onset diarrhea. In total, 108 patients from 15 institutions were enrolled in this study. We collected clinical data from all patients. Seventy-three patients with exudative diarrhea, 4 with osmotic or secretory diarrhea and 8 with protein-losing enteropathy were subjected to genetic analysis. RESULTS A total of 15 out of the 108 enrolled patients (13.9%) were identified as monogenic. We identified 1 patient with RELA, 2 with TNFAIP3, 1 with CTLA4, 1 with SLCO2A1, 4 with XIAP, 3 with IL10RA, 1 with HPS1, 1 with FOXP3, and 1 with CYBB gene mutations. We also identified 1 patient with NFKB2 and 1 with TERT mutations from the gene panel for primary immunodeficiency syndromes. The patient with refractory diarrhea caused by heterozygous truncated RelA protein expression is the first case identified worldwide, and functional analysis revealed that the mutation affected nuclear factor kappa B signaling. Genotypes were significantly associated with the clinical and pathological findings in each patient. CONCLUSIONS We identified variable monogenic diseases in the patients and found that genes responsible for primary immunodeficiency diseases were frequently involved in molecular pathogenesis. Comprehensive genetic analysis was useful for accurate molecular diagnosis, understanding of underlying pathogenesis, and selecting the optimal treatment for patients with early-onset refractory diarrhea.An infographic for this article is available at: http://links.lww.com/MPG/B853.
Collapse
|
14
|
de Gregorio E, Colell A, Morales A, Marí M. Relevance of SIRT1-NF-κB Axis as Therapeutic Target to Ameliorate Inflammation in Liver Disease. Int J Mol Sci 2020; 21:E3858. [PMID: 32485811 PMCID: PMC7312021 DOI: 10.3390/ijms21113858] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an adaptive response in pursuit of homeostasis reestablishment triggered by harmful conditions or stimuli, such as an infection or tissue damage. Liver diseases cause approximately 2 million deaths per year worldwide and hepatic inflammation is a common factor to all of them, being the main driver of hepatic tissue damage and causing progression from non-alcoholic fatty liver disease (NAFLD) to non-alcoholic steatohepatitis (NASH), cirrhosis and, ultimately, hepatocellular carcinoma (HCC). The metabolic sensor SIRT1, a class III histone deacetylase with strong expression in metabolic tissues such as the liver, and transcription factor NF-κB, a master regulator of inflammatory response, show an antagonistic relationship in controlling inflammation. For this reason, SIRT1 targeting is emerging as a potential strategy to improve different metabolic and/or inflammatory pathologies. In this review, we explore diverse upstream regulators and some natural/synthetic activators of SIRT1 as possible therapeutic treatment for liver diseases.
Collapse
Affiliation(s)
- Estefanía de Gregorio
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, 08036 Barcelona, Spain;
| | - Anna Colell
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain;
| | - Albert Morales
- Department of Cell Death and Proliferation, IIBB-CSIC, Barcelona Clinic Liver Cancer Group, Liver Unit, Hospital Clínic of Barcelona, University of Barcelona, CIBEREHD, 08036 Barcelona, Spain;
| | - Montserrat Marí
- Department of Cell Death and Proliferation, IIBB-CSIC, IDIBAPS, 08036 Barcelona, Spain;
| |
Collapse
|
15
|
Ouyang G, Liao Q, Zhang D, Rong F, Cai X, Fan S, Zhu J, Wang J, Liu X, Liu X, Xiao W. Zebrafish NF-κB/p65 Is Required for Antiviral Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:3019-3029. [PMID: 32321758 DOI: 10.4049/jimmunol.1900309] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 03/30/2020] [Indexed: 01/08/2023]
Abstract
Transcriptional programs regulated by the NF-κB family are essential for the inflammatory response as well as for innate and adaptive immunity. NF-κB activation occurs via two major signaling pathways: the canonical and the noncanonical. The canonical NF-κB pathway responds to diverse immune stimulations and leads to rapid but transient activation. As a member of the canonical NF-κB family, p65 is thought to be a key regulator of viral infection. Because of the embryonic lethality of p65-null mice, the physiological role of p65 in the antiviral immune response is still unclear. In this study, we generated p65-null zebrafish, which were viable and indistinguishable from their wildtype (WT) siblings under normal conditions. However, p65-null zebrafish were more sensitive to spring viremia of carp virus infection than their WT siblings. Further assays indicated that proinflammatory and antiviral genes, including IFN, were downregulated in p65-null zebrafish after spring viremia of carp virus infection compared with their WT siblings. Our results thus suggested that p65 is required for the antiviral response, activating not only proinflammatory genes but also antiviral genes (including IFN).
Collapse
Affiliation(s)
- Gang Ouyang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Qian Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Dawei Zhang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Fangjing Rong
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xiaolian Cai
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Sijia Fan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Junji Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xing Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China.,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| | - Xueqin Liu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Wuhan Xiao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China; .,The Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Wuhan 430072, China.,The Key Laboratory of Aquatic Biodiversity and Conservation, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, People's Republic of China.,The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan 430072, China.,University of Chinese Academy of Sciences, Beijing 100049, China; and
| |
Collapse
|
16
|
Jarosz-Griffiths HH, Holbrook J, Lara-Reyna S, McDermott MF. TNF receptor signalling in autoinflammatory diseases. Int Immunol 2020; 31:639-648. [PMID: 30838383 DOI: 10.1093/intimm/dxz024] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 03/01/2019] [Indexed: 12/16/2022] Open
Abstract
Autoinflammatory syndromes are a group of disorders characterized by recurring episodes of inflammation as a result of specific defects in the innate immune system. Patients with autoinflammatory disease present with recurrent outbreaks of chronic systemic inflammation that are mediated by innate immune cells, for the most part. A number of these diseases arise from defects in the tumour necrosis factor receptor (TNFR) signalling pathway leading to elevated levels of inflammatory cytokines. Elucidation of the molecular mechanisms of these recently defined autoinflammatory diseases has led to a greater understanding of the mechanisms of action of key molecules involved in TNFR signalling, particularly those involved in ubiquitination, as found in haploinsufficiency of A20 (HA20), otulipenia/OTULIN-related autoinflammatory syndrome (ORAS) and linear ubiquitin chain assembly complex (LUBAC) deficiency. In this review, we also address other TNFR signalling disorders such as TNFR-associated periodic syndrome (TRAPS), RELA haploinsufficiency, RIPK1-associated immunodeficiency and autoinflammation, X-linked ectodermal dysplasia and immunodeficiency (X-EDA-ID) and we review the most recent advances surrounding these diseases and therapeutic approaches currently used to target these diseases. Finally, we explore therapeutic advances in TNF-related immune-based therapies and explore new approaches to target disease-specific modulation of autoinflammatory diseases.
Collapse
Affiliation(s)
- Heledd H Jarosz-Griffiths
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, University of Leeds, Leeds, UK.,Leeds Institute of Medical Research at St James's, Clinical Sciences Building, University of Leeds, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Wellcome Trust Brenner Building, University of Leeds, Leeds, UK
| | - Jonathan Holbrook
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, University of Leeds, Leeds, UK.,Leeds Institute of Medical Research at St James's, Clinical Sciences Building, University of Leeds, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Wellcome Trust Brenner Building, University of Leeds, Leeds, UK
| | - Samuel Lara-Reyna
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, University of Leeds, Leeds, UK.,Leeds Institute of Medical Research at St James's, Clinical Sciences Building, University of Leeds, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Wellcome Trust Brenner Building, University of Leeds, Leeds, UK
| | - Michael F McDermott
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), Wellcome Trust Brenner Building, University of Leeds, Leeds, UK.,Leeds Cystic Fibrosis Trust Strategic Research Centre, Wellcome Trust Brenner Building, University of Leeds, Leeds, UK
| |
Collapse
|
17
|
Lambrou GI, Hatziagapiou K, Vlahopoulos S. Inflammation and tissue homeostasis: the NF-κB system in physiology and malignant progression. Mol Biol Rep 2020; 47:4047-4063. [PMID: 32239468 DOI: 10.1007/s11033-020-05410-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Disruption of tissue function activates cellular stress which triggers a number of mechanisms that protect the tissue from further damage. These mechanisms involve a number of homeostatic modules, which are regulated at the level of gene expression by the transactivator NF-κB. This transcription factor shifts between activation and repression of discrete, cell-dependent gene expression clusters. Some of its target genes provide feedback to NF-κB itself, thereby strengthening the inflammatory response of the tissue and later terminating inflammation to facilitate restoration of tissue homeostasis. Disruption of key feedback modules for NF-κB in certain cell types facilitates the survival of clones with genomic aberrations, and protects them from being recognized and eliminated by the immune system, to enable thereby carcinogenesis.
Collapse
Affiliation(s)
- George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece.
| |
Collapse
|
18
|
Farabaugh KT, Krokowski D, Guan BJ, Gao Z, Gao XH, Wu J, Jobava R, Ray G, de Jesus TJ, Bianchi MG, Chukwurah E, Bussolati O, Kilberg M, Buchner DA, Sen GC, Cotton C, McDonald C, Longworth M, Ramakrishnan P, Hatzoglou M. PACT-mediated PKR activation acts as a hyperosmotic stress intensity sensor weakening osmoadaptation and enhancing inflammation. eLife 2020; 9:e52241. [PMID: 32175843 PMCID: PMC7145421 DOI: 10.7554/elife.52241] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 03/14/2020] [Indexed: 12/15/2022] Open
Abstract
The inability of cells to adapt to increased environmental tonicity can lead to inflammatory gene expression and pathogenesis. The Rel family of transcription factors TonEBP and NF-κB p65 play critical roles in the switch from osmoadaptive homeostasis to inflammation, respectively. Here we identified PACT-mediated PKR kinase activation as a marker of the termination of adaptation and initiation of inflammation in Mus musculus embryonic fibroblasts. We found that high stress-induced PACT-PKR activation inhibits the interaction between NF-κB c-Rel and TonEBP essential for the increased expression of TonEBP-dependent osmoprotective genes. This resulted in enhanced formation of TonEBP/NF-κB p65 complexes and enhanced proinflammatory gene expression. These data demonstrate a novel role of c-Rel in the adaptive response to hyperosmotic stress, which is inhibited via a PACT/PKR-dependent dimer redistribution of the Rel family transcription factors. Our results suggest that inhibiting PACT-PKR signaling may prove a novel target for alleviating stress-induced inflammatory diseases.
Collapse
Affiliation(s)
- Kenneth T Farabaugh
- Department of Pharmacology, Case Western Reserve UniversityClevelandUnited States
| | - Dawid Krokowski
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
- Department of Molecular Biology, Maria Curie-Sklodowska UniversityLublinPoland
| | - Bo-Jhih Guan
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| | - Zhaofeng Gao
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| | - Xing-Huang Gao
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| | - Jing Wu
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| | - Raul Jobava
- Department of Biochemistry, Case Western Reserve UniversityClevelandUnited States
| | - Greeshma Ray
- Department of Inflammation and Immunity, Cleveland Clinic FoundationClevelandUnited States
| | - Tristan J de Jesus
- Department of Pathology, Case Western Reserve UniversityClevelandUnited States
| | | | - Evelyn Chukwurah
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| | - Ovidio Bussolati
- Department of Medicine and Surgery, Universita degli Studi di ParmaParmaItaly
| | - Michael Kilberg
- Department of Biochemistry and Molecular Biology, University of FloridaGainesvilleUnited States
| | - David A Buchner
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
- Department of Biochemistry, Case Western Reserve UniversityClevelandUnited States
| | - Ganes C Sen
- Department of Inflammation and Immunity, Cleveland Clinic FoundationClevelandUnited States
| | - Calvin Cotton
- Department of Physiology and Biophysics, Case Western Reserve UniversityClevelandUnited States
| | - Christine McDonald
- Department of Inflammation and Immunity, Cleveland Clinic FoundationClevelandUnited States
| | - Michelle Longworth
- Department of Inflammation and Immunity, Cleveland Clinic FoundationClevelandUnited States
| | | | - Maria Hatzoglou
- Department of Genetics and Genome Sciences, Case Western Reserve UniversityClevelandUnited States
| |
Collapse
|
19
|
Abstract
Genetic robustness, or the ability of an organism to maintain fitness in the presence of mutations, can be achieved via protein feedback loops. Recent evidence suggests that organisms may also respond to mutations by upregulating related gene(s) independently of protein feedback loops, a phenomenon called transcriptional adaptation. However, the prevalence of transcriptional adaptation and its underlying molecular mechanisms are unknown. Here, by analyzing several models of transcriptional adaptation in zebrafish and mouse, we show a requirement for mRNA degradation. Alleles that fail to transcribe the mutated gene do not display transcriptional adaptation and exhibit more severe phenotypes than alleles displaying mutant mRNA decay. Transcriptome analysis reveals the upregulation of a substantial proportion of the genes that exhibit sequence similarity with the mutated gene’s mRNA, suggesting a sequence dependent mechanism. Besides implications for our understanding of disease-causing mutations, these findings will help design mutant alleles with minimal transcriptional adaptation-derived compensation.
Collapse
|
20
|
Verma P, Verma R, Nair RR, Budhwar S, Khanna A, Agrawal NR, Sinha R, Birendra R, Rajender S, Singh K. Altered crosstalk of estradiol and progesterone with Myeloid-derived suppressor cells and Th1/Th2 cytokines in early miscarriage is associated with early breakdown of maternal-fetal tolerance. Am J Reprod Immunol 2019; 81:e13081. [PMID: 30589483 DOI: 10.1111/aji.13081] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 11/30/2018] [Accepted: 12/20/2018] [Indexed: 12/28/2022] Open
Abstract
PROBLEM Decline in myeloid-derived suppressor cells (MDSCs) and Th2 cytokines levels lead to early miscarriage (EM) but how the hormonal milieu of the body regulates MDSCs and Th1/Th2 cytokine balance is still a matter of investigation. METHOD OF STUDY Peripheral blood and decidua samples were collected from 20 EM patients, and 20 healthy pregnant women opted for elective abortion. MDSCs and G-MDSCs levels were analyzed in peripheral blood mononuclear cells, and Th1/Th2 cytokines levels were determined in serum via flow cytometry. Estrogen (E2), Progesterone (P4), and Testosterone levels were measured via ELISA. Further, proliferation and apoptosis in decidual samples were checked via immunoblot/immunohistochemistry of estrogen receptor -α (ER-α), STAT-3/pSTAT-3, and caspase-3, respectively. RESULTS Our results clearly indicate that in EM patients; decline in E2 and P4 significantly correlates with decline in MDSCs, particularly with subtype granulocytic MDSCs (G-MDSCs) and skewness of the Th1/Th2 cytokines balance toward Th1 response. Downregulation of ER- α and increased caspase-3 expression in endometrium decidua signifies poor endometrial receptivity in EM. STAT-3 activation regulates proliferation, differentiation and suppressive potency of MDSCs. In decidua of EM, significantly lower expression of pSTAT-3 indicates that these processes pertaining to MDSCs are compromised. CONCLUSION Altogether, this unfavorable systemic milieu may drive toward early breakdown of maternal-fetal tolerance in EM. Therefore, regulated crosstalk of E2, P4 with MDSCs and balanced Th1/Th2 cytokines is prerequisite for successful pregnancy.
Collapse
Affiliation(s)
- Priyanka Verma
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| | - Rachna Verma
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| | - Rohini R Nair
- Department of Molecular & Human Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Snehil Budhwar
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| | - Anuradha Khanna
- Department of Obstetrics & Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Nisha Rani Agrawal
- Department of Obstetrics & Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ruchi Sinha
- Department of Obstetrics & Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ruchi Birendra
- Department of Obstetrics & Gynecology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | | | - Kiran Singh
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| |
Collapse
|
21
|
Concetti J, Wilson CL. NFKB1 and Cancer: Friend or Foe? Cells 2018; 7:cells7090133. [PMID: 30205516 PMCID: PMC6162711 DOI: 10.3390/cells7090133] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 12/30/2022] Open
Abstract
Current evidence strongly suggests that aberrant activation of the NF-κB signalling pathway is associated with carcinogenesis. A number of key cellular processes are governed by the effectors of this pathway, including immune responses and apoptosis, both crucial in the development of cancer. Therefore, it is not surprising that dysregulated and chronic NF-κB signalling can have a profound impact on cellular homeostasis. Here we discuss NFKB1 (p105/p50), one of the five subunits of NF-κB, widely implicated in carcinogenesis, in some cases driving cancer progression and in others acting as a tumour-suppressor. The complexity of the role of this subunit lies in the multiple dimeric combination possibilities as well as the different interacting co-factors, which dictate whether gene transcription is activated or repressed, in a cell and organ-specific manner. This review highlights the multiple roles of NFKB1 in the development and progression of different cancers, and the considerations to make when attempting to manipulate NF-κB as a potential cancer therapy.
Collapse
Affiliation(s)
- Julia Concetti
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear NE2 4HH, UK.
| | - Caroline L Wilson
- Newcastle Fibrosis Research Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, Tyne and Wear NE2 4HH, UK.
| |
Collapse
|
22
|
Abstract
This review by Shan et al. discusses necroptosis, a form of regulated necrotic cell death mediated by RIPK1 kinase activity, RIPK3, and MLKL, which can be activated under apoptosis-deficient conditions. Both necroptosis and apoptosis can be activated in response to various mutations that result in the abortion of defective embryos and during human inflammatory and neurodegenerative pathologies. Necroptosis, a form of regulated necrotic cell death mediated by RIPK1 (receptor-interacting protein kinase 1) kinase activity, RIPK3, and MLKL (mixed-lineage kinase domain-like pseudokinase), can be activated under apoptosis-deficient conditions. Modulating the activation of RIPK1 by ubiquitination and phosphorylation is critical to control both necroptosis and apoptosis. Mutant mice with kinase-dead RIPK1 or RIPK3 and MLKL deficiency show no detrimental phenotype in regard to development and adult homeostasis. However, necroptosis and apoptosis can be activated in response to various mutations that result in the abortion of the defective embryos and human inflammatory and neurodegenerative pathologies. RIPK1 inhibition represents a key therapeutic strategy for treatment of diseases where blocking both necroptosis and apoptosis can be beneficial.
Collapse
Affiliation(s)
- Bing Shan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China
| | - Heling Pan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China
| | - Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, PuDong District, Shanghai 201203, China.,Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
23
|
Feketeova E, Li Z, Joseph B, Shah R, Spolarics Z, Ulloa L. Dopaminergic Control of Inflammation and Glycemia in Sepsis and Diabetes. Front Immunol 2018; 9:943. [PMID: 29780390 PMCID: PMC5945822 DOI: 10.3389/fimmu.2018.00943] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 04/16/2018] [Indexed: 12/21/2022] Open
Abstract
Most preclinical treatments for sepsis failed in clinical trials in part because the experimental models of sepsis were performed on healthy animals that do not mimic septic patients. Here, we report that experimental diabetes worsens glycemia, inflammation, and mortality in experimental sepsis. Diabetes increases hyperglycemia, systemic inflammation, and mortality in sepsis. Diabetes exacerbates serum tumor necrosis factor (TNF) levels in sepsis by increasing splenic TNF production. Both serum from diabetic mice and glucose increase cytokine production in splenocytes. Anti-inflammatory treatments cannot control hyperglycemia and are less effective in diabetic patients. By contrast, dopaminergic agonist type-1, fenoldopam, attenuates hyperglycemia, and systemic inflammation in diabetic septic mice by inhibiting splenic p65NF-kB phosphorylation. Fenoldopam inhibits TNF production in splenocytes even at high glucose concentrations and inhibits the canonical NF-kB pathway by inhibiting p65RelA and p50NF-kB1 phosphorylation without affecting the non-canonical NF-kB proteins. Treatment with fenoldopam rescues diabetic mice from established polymicrobial peritonitis even when the treatment is started after the onset of sepsis. These results suggest that dopaminergic agonists can control hyperglycemia, systemic inflammation and provide therapeutic advantages for treating diabetic patients with sepsis in a clinically relevant time frame.
Collapse
Affiliation(s)
- Eleonora Feketeova
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Zhifeng Li
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Biju Joseph
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Roshan Shah
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Zoltan Spolarics
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| | - Luis Ulloa
- Department of Surgery, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Newark, NJ, United States
| |
Collapse
|
24
|
Xu C, Wu X, Zhang X, Xie Q, Fan C, Zhang H. Embryonic Lethality and Host Immunity of RelA-Deficient Mice Are Mediated by Both Apoptosis and Necroptosis. THE JOURNAL OF IMMUNOLOGY 2017; 200:271-285. [PMID: 29167229 DOI: 10.4049/jimmunol.1700859] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 10/26/2017] [Indexed: 01/10/2023]
Abstract
In mammalian cells, signaling pathways triggered by TNF can be switched from NF-κB activation to apoptosis and/or necroptosis. The in vivo mechanisms underlying the mutual regulation of these three signaling pathways are poorly understood. In this article, we report that the embryonic lethality of RelA-deficient mice is partially prevented by the deletion of Rip3 or Mlkl, but it is fully rescued by the combined ablation of Fadd and Rip3 or Mlkl or by blocking RIP1 kinase activity (RIP1K45A). RelA-/-Fadd-/-Rip3-/- triple-knockout (TKO) and RelA-/-Rip1K45A/K45A mice displayed bacterial pneumonia leading to death ∼2 wk after birth. Moreover, RelA-/-Rip1K45A/K45A mice, but not TKO mice, developed severe inflammation associated with inflammatory skin lesion. Antibiotic treatment improved bacterial pneumonia, extended the lifespan of TKO and RelA-/-Rip1K45A/K45A mice, and alleviated skin inflammation in RelA-/-Rip1K45A/K45A mice. These results show the mechanisms underlying the in vivo mutual regulation between NF-κB activation and the cell death pathway and provide new insights into this interplay in embryonic development and host immune homeostasis.
Collapse
Affiliation(s)
- Chengxian Xu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Xiaoxia Wu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Xixi Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Qun Xie
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and.,Department of Anesthesiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Cunxian Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| | - Haibing Zhang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; and
| |
Collapse
|
25
|
Mitchell JP, Carmody RJ. NF-κB and the Transcriptional Control of Inflammation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 335:41-84. [PMID: 29305014 DOI: 10.1016/bs.ircmb.2017.07.007] [Citation(s) in RCA: 342] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The NF-κB transcription factor was discovered 30 years ago and has since emerged as the master regulator of inflammation and immune homeostasis. It achieves this status by means of the large number of important pro- and antiinflammatory factors under its transcriptional control. NF-κB has a central role in inflammatory diseases such as rheumatoid arthritis, inflammatory bowel disease, and autoimmunity, as well as diseases comprising a significant inflammatory component such as cancer and atherosclerosis. Here, we provide an overview of the studies that form the basis of our understanding of the role of NF-κB subunits and their regulators in controlling inflammation. We also describe the emerging importance of posttranslational modifications of NF-κB in the regulation of inflammation, and highlight the future challenges faced by researchers who aim to target NF-κB transcriptional activity for therapeutic benefit in treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jennifer P Mitchell
- Rheumatoid Arthritis Pathogenesis Centre of Excellence, Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Ruaidhrí J Carmody
- Centre for Immunobiology, Institute of Infection, Immunity, and Inflammation, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
26
|
Qi R, Huang J, Wang Q, Liu H, Wang R, Wang J, Yang F. MicroRNA‐224‐5p regulates adipocyte apoptosis induced by TNFα via controlling NF‐κB activation. J Cell Physiol 2017; 233:1236-1246. [DOI: 10.1002/jcp.25992] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Renli Qi
- Chongqing Academy of Animal ScienceRongchangChongqingChina
- Key Laboratory of Pig Industry SciencesMinistry of AgricultureRongchangChongqingChina
| | - Jinxiu Huang
- Chongqing Academy of Animal ScienceRongchangChongqingChina
- Key Laboratory of Pig Industry SciencesMinistry of AgricultureRongchangChongqingChina
| | - Qi Wang
- Chongqing Academy of Animal ScienceRongchangChongqingChina
| | - Hong Liu
- Chongqing Academy of Animal ScienceRongchangChongqingChina
| | - Ruisheng Wang
- Key Laboratory of Pig Industry SciencesMinistry of AgricultureRongchangChongqingChina
| | - Jing Wang
- Chongqing Academy of Animal ScienceRongchangChongqingChina
| | - Feiyun Yang
- Chongqing Academy of Animal ScienceRongchangChongqingChina
| |
Collapse
|
27
|
Bettermann K. NF-κB and Its Implication in Liver Health and Cancer Development. MECHANISMS OF MOLECULAR CARCINOGENESIS – VOLUME 1 2017:87-114. [DOI: 10.1007/978-3-319-53659-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Ijaz T, Wakamiya M, Sun H, Recinos A, Tilton RG, Brasier AR. Generation and characterization of a novel transgenic mouse harboring conditional nuclear factor-kappa B/RelA knockout alleles. BMC DEVELOPMENTAL BIOLOGY 2016; 16:32. [PMID: 27662828 PMCID: PMC5034478 DOI: 10.1186/s12861-016-0135-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022]
Abstract
Background Nuclear Factor-Kappa B (NF-kB) is a family of transcription factors that are important in embryonic development, inflammation, epithelial-to-mesenchymal transition and cancer. The 65 kDa RelA subunit is the major transcriptional activator of the NF-kB pathways. Whole-body deficiency of RelA leads to massive apoptosis of liver hepatocytes and death in utero. To study the role of RelA in physiology and in disease states in a manner that circumvents this embryonic lethal phenotype, we have generated a mouse with RelA conditional knockout (CKO) alleles containing loxP sites that are deleted by activated Cre recombinase. Results We demonstrate that RelACKO/CKO mice are fertile, do not display any developmental defects and can be crossed with Cre-expressing mice to delete RelA in a temporal, tissue-specific manner. Our mating of RelACKO/CKO mice with Zp3-Cre transgenic led to embryonic lethality of RelA-deficient embryos. In contrast, mating of RelACKO/CKO mice with Col1α2-CreER mice allowed for the generation of double transgenics which could be stimulated with tamoxifen to induce fibroblast-specific RelA deletion in adulthood. Conclusions Based on our collective data, we conclude that this novel RelACKO/CKO mouse allows for efficient deletion of RelA in a tissue-specific manner. This RelACKO/CKO mouse will be an invaluable tool for deciphering the mechanistic roles of RelA in various cells and tissues during development and in disease. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0135-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Talha Ijaz
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maki Wakamiya
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA
| | - Hong Sun
- Internal Medicine-Division of Endocrinology, University of Texas Medical Branch, Galveston, TX, USA
| | - Adrian Recinos
- Internal Medicine-Division of Endocrinology, University of Texas Medical Branch, Galveston, TX, USA
| | - Ronald G Tilton
- Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA.,Internal Medicine-Division of Endocrinology, University of Texas Medical Branch, Galveston, TX, USA.,Sealy Center for Molecular Medicine, University of Texas Medical Branch, MRB 8.128, 301 University Blvd, Galveston, TX, 77555-1060, USA
| | - Allan R Brasier
- Departments of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA. .,Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, USA. .,Internal Medicine-Division of Endocrinology, University of Texas Medical Branch, Galveston, TX, USA. .,Sealy Center for Molecular Medicine, University of Texas Medical Branch, MRB 8.128, 301 University Blvd, Galveston, TX, 77555-1060, USA.
| |
Collapse
|
29
|
Espín-Palazón R, Traver D. The NF-κB family: Key players during embryonic development and HSC emergence. Exp Hematol 2016; 44:519-27. [PMID: 27132652 DOI: 10.1016/j.exphem.2016.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 03/24/2016] [Accepted: 03/26/2016] [Indexed: 02/07/2023]
Abstract
The nuclear factor-κB (NF-κB) family is a crucial transcription factor group known mainly for its role in the regulation of the immune system and its response to infection in vertebrates. The signaling pathway leading to NF-κB activation and translocation to the nucleus to exert its function as a transcription factor is well conserved among Kingdom Animalia, which has helped to elucidate other roles that NF-κB plays in other biological contexts such as developmental biology. The manipulation of NF-κB members in a diverse range of animal models results in severe developmental defects during embryogenesis, very often leading to embryonic lethality. Defects include dorsal-ventral patterning and limb, liver, skin, lung, neural, notochord, muscle, skeletal, and hematopoietic defects. Here, we recapitulate the research that has been done to address the role that NF-κB plays during embryonic development, in particular to emphasize its recently discovered role in the specification of hematopoietic stem cells (HSCs), the foundation of the hematopoietic system in vertebrates.
Collapse
Affiliation(s)
- Raquel Espín-Palazón
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA
| | - David Traver
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA.
| |
Collapse
|
30
|
An NF-κB--EphrinA5-Dependent Communication between NG2(+) Interstitial Cells and Myoblasts Promotes Muscle Growth in Neonates. Dev Cell 2016; 36:215-24. [PMID: 26777211 DOI: 10.1016/j.devcel.2015.12.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 11/04/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023]
Abstract
Skeletal muscle growth immediately following birth is critical for proper body posture and locomotion. However, compared with embryogenesis and adulthood, the processes regulating the maturation of neonatal muscles is considerably less clear. Studies in the 1960s predicted that neonatal muscle growth results from nuclear accretion of myoblasts preferentially at the tips of myofibers. Remarkably, little information has been added since then to resolve how myoblasts migrate to the ends of fibers. Here, we provide insight into this process by revealing a unique NF-κB-dependent communication between NG2(+) interstitial cells and myoblasts. NF-κB in NG2(+) cells promotes myoblast migration to the tips of myofibers through cell-cell contact. This occurs through expression of ephrinA5 from NG2(+) cells, which we further deduce is an NF-κB target gene. Together, these results suggest that NF-κB plays an important role in the development of newborn muscles to ensure proper myoblast migration for fiber growth.
Collapse
|
31
|
Hilliard KL, Allen E, Traber KE, Yamamoto K, Stauffer NM, Wasserman GA, Jones MR, Mizgerd JP, Quinton LJ. The Lung-Liver Axis: A Requirement for Maximal Innate Immunity and Hepatoprotection during Pneumonia. Am J Respir Cell Mol Biol 2015; 53:378-90. [PMID: 25607543 DOI: 10.1165/rcmb.2014-0195oc] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The hepatic acute-phase response (APR), stimulated by injury or inflammation, is characterized by significant changes in circulating acute-phase protein (APP) concentrations. Although individual functions of liver-derived APPs are known, the net consequence of APP changes is unclear. Pneumonia, which induces the APR, causes an inflammatory response within the airspaces that is coordinated largely by alveolar macrophages and is typified by cytokine production, leukocyte recruitment, and plasma extravasation, the latter of which may enable delivery of hepatocyte-derived APPs to the infection site. To determine the functional significance of the hepatic APR during pneumonia, we challenged APR-null mice lacking hepatocyte signal transducer and activator of transcription 3 (STAT3) and v-rel avian reticuloendotheliosis viral oncogene homolog A (RelA) with Escherichia coli in the airspaces. APR-null mice displayed ablated APP induction, significantly increased mortality, liver injury and apoptosis, and a trend toward increased bacterial burdens. TNF-α neutralization reversed hepatotoxicity, but not mortality, suggesting that APR-dependent survival is not solely due to hepatoprotection. After a milder (nonlethal) E. coli infection, hepatocyte-specific mutations decreased APP concentrations and pulmonary inflammation in bronchoalveolar lavage fluid. Cytokine expression in airspace macrophages, but not other airspace or circulating cells, was significantly dependent on APP extravasation into the alveoli. These data identify a novel signaling axis whereby the liver response enhances macrophage activation and pulmonary inflammation during pneumonia. Although hepatic acute-phase changes directly curb injury induced by TNF-α in the liver itself, APPs downstream of these same signals promote survival in association with innate immunity in the lungs, thus demonstrating a critical role for the lung-liver axis during pneumonia.
Collapse
Affiliation(s)
- Kristie L Hilliard
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Eri Allen
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Katrina E Traber
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Kazuko Yamamoto
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Nicole M Stauffer
- 2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Gregory A Wasserman
- Departments of 1 Microbiology.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Matthew R Jones
- 3 Medicine.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Joseph P Mizgerd
- Departments of 1 Microbiology.,3 Medicine.,4 Biochemistry, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| | - Lee J Quinton
- 3 Medicine.,5 Pathology and Laboratory Medicine, and.,2 Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
32
|
Lopez MA, Meier D, Wong WWL, Fontana A. TNF induced inhibition of Cirbp expression depends on RelB NF-κB signalling pathway. Biochem Biophys Rep 2015; 5:22-26. [PMID: 28955803 PMCID: PMC5600431 DOI: 10.1016/j.bbrep.2015.11.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 10/27/2015] [Accepted: 11/09/2015] [Indexed: 12/27/2022] Open
Abstract
The circadian clock is required for the rhythmic expression of a plethora of genes that orchestrate metabolism, sleep-wake behaviour and the immune response to pathogens. The cold-inducible RNA binding protein (CIRBP) is required for high amplitude expression of clock genes. Moreover, CIRBP protects the expression of clock genes from the inhibitory effects of tumour necrosis factor (TNF). However, since TNF represses Cirbp expression, the protective effect of CIRBP is lost. Here, we show that the TNF effect on Cirbp requires the non-canonical NF-κB signalling pathway. While a knock down of RelA does not alter the effects of TNF on Cirbp, a knock down of RelB represses this effect. In addition, the data indicate that p50 and p52 are required in the TNF induced inhibition of Cirbp. These results show that Cirbp expression in TNF treated cells is regulated via the non-canonical NF-κB pathway.
Collapse
Affiliation(s)
- Martin A Lopez
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Daniel Meier
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - W Wei-Lynn Wong
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| | - Adriano Fontana
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
33
|
Zhao Y, Ma CA, Wu L, Iwai K, Ashwell JD, Oltz EM, Ballard DW, Jain A. CYLD and the NEMO Zinc Finger Regulate Tumor Necrosis Factor Signaling and Early Embryogenesis. J Biol Chem 2015. [PMID: 26224629 DOI: 10.1074/jbc.m115.658096] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NF-κB essential modulator (NEMO) and cylindromatosis protein (CYLD) are intracellular proteins that regulate the NF-κB signaling pathway. Although mice with either CYLD deficiency or an alteration in the zinc finger domain of NEMO (K392R) are born healthy, we found that the combination of these two gene defects in double mutant (DM) mice is early embryonic lethal but can be rescued by the absence of TNF receptor 1 (TNFR1). Notably, NEMO was not recruited into the TNFR1 complex of DM cells, and consequently NF-κB induction by TNF was severely impaired and DM cells were sensitized to TNF-induced cell death. Interestingly, the TNF signaling defects can be fully rescued by reconstitution of DM cells with CYLD lacking ubiquitin hydrolase activity but not with CYLD mutated in TNF receptor-associated factor 2 (TRAF2) or NEMO binding sites. Therefore, our data demonstrate an unexpected non-catalytic function for CYLD as an adapter protein between TRAF2 and the NEMO zinc finger that is important for TNF-induced NF-κB signaling during embryogenesis.
Collapse
Affiliation(s)
- Yongge Zhao
- From the Laboratory of Host Defenses, NIAID, National Institutes of Health, Bethesda, Maryland 20892,
| | - Chi A Ma
- From the Laboratory of Host Defenses, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Liming Wu
- From the Laboratory of Host Defenses, NIAID, National Institutes of Health, Bethesda, Maryland 20892
| | - Kazuhiro Iwai
- the Department of Molecular and Cellular Physiology, Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Jonathan D Ashwell
- the Laboratory of Immune Cell Biology, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Eugene M Oltz
- the Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110, and
| | - Dean W Ballard
- the Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, Tennessee 37232
| | - Ashish Jain
- From the Laboratory of Host Defenses, NIAID, National Institutes of Health, Bethesda, Maryland 20892,
| |
Collapse
|
34
|
Fukazawa T, Hiraiwa N, Umemura T, Mise-Omata S, Obata Y, Doi T. Egress of Mature Murine Regulatory T Cells from the Thymus Requires RelA. THE JOURNAL OF IMMUNOLOGY 2015; 194:3020-8. [DOI: 10.4049/jimmunol.1302756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
35
|
Nair RR, Khanna A, Singh K. Association of increased S100A8 serum protein with early pregnancy loss. Am J Reprod Immunol 2014; 73:91-4. [PMID: 25252120 DOI: 10.1111/aji.12318] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 08/18/2014] [Indexed: 12/20/2022] Open
Abstract
PROBLEM The contribution of systemic S100A8 protein in menstrual cycle, pregnancy, and early pregnancy loss (EPL) is not known. Altered expression of S100A8 in maternal decidua is associated with recurrent early pregnancy loss. The objective of this study was to investigate the systemic level of S100A8 in different phases of menstrual cycle, different trimester of pregnancy, and in EPL. METHOD OF STUDY Level of S100A8 was investigated in serum samples of the subjects through enzyme-linked immunosorbent assay (ELISA). RESULT AND CONCLUSION S100A8 levels were elevated during proliferative phase of menstrual cycle. We found no statistical difference in S100A8 level in different trimester of pregnancy. S100A8 level was found to be significantly elevated in patients with EPL. This is the first study evaluating the systemic level of S100A8 predicting its role during menstrual cycle and pregnancy. It opens a new perspective in which S100A8 can be used as a prognostic marker for EPL.
Collapse
Affiliation(s)
- Rohini R Nair
- Department of Molecular & Human Genetics, Banaras Hindu University, Varanasi, India
| | | | | |
Collapse
|
36
|
Ehlken H, Krishna-Subramanian S, Ochoa-Callejero L, Kondylis V, Nadi NE, Straub BK, Schirmacher P, Walczak H, Kollias G, Pasparakis M. Death receptor-independent FADD signalling triggers hepatitis and hepatocellular carcinoma in mice with liver parenchymal cell-specific NEMO knockout. Cell Death Differ 2014; 21:1721-32. [PMID: 24971483 DOI: 10.1038/cdd.2014.83] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 05/02/2014] [Accepted: 05/06/2014] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) usually develops in the context of chronic hepatitis triggered by viruses or toxic substances causing hepatocyte death, inflammation and compensatory proliferation of liver cells. Death receptors of the TNFR superfamily regulate cell death and inflammation and are implicated in liver disease and cancer. Liver parenchymal cell-specific ablation of NEMO/IKKγ, a subunit of the IκB kinase (IKK) complex that is essential for the activation of canonical NF-κB signalling, sensitized hepatocytes to apoptosis and caused the spontaneous development of chronic hepatitis and HCC in mice. Here we show that hepatitis and HCC development in NEMO(LPC-KO) mice is triggered by death receptor-independent FADD-mediated hepatocyte apoptosis. TNF deficiency in all cells or conditional LPC-specific ablation of TNFR1, Fas or TRAIL-R did not prevent hepatocyte apoptosis, hepatitis and HCC development in NEMO(LPC-KO) mice. To address potential functional redundancies between death receptors we generated and analysed NEMO(LPC-KO) mice with combined LPC-specific deficiency of TNFR1, Fas and TRAIL-R and found that also simultaneous lack of all three death receptors did not prevent hepatocyte apoptosis, chronic hepatitis and HCC development. However, LPC-specific combined deficiency in TNFR1, Fas and TRAIL-R protected the NEMO-deficient liver from LPS-induced liver failure, showing that different mechanisms trigger spontaneous and LPS-induced hepatocyte apoptosis in NEMO(LPC-KO) mice. In addition, NK cell depletion did not prevent liver damage and hepatitis. Moreover, NEMO(LPC-KO) mice crossed into a RAG-1-deficient genetic background-developed hepatitis and HCC. Collectively, these results show that the spontaneous development of hepatocyte apoptosis, chronic hepatitis and HCC in NEMO(LPC-KO) mice occurs independently of death receptor signalling, NK cells and B and T lymphocytes, arguing against an immunological trigger as the critical stimulus driving hepatocarcinogenesis in this model.
Collapse
Affiliation(s)
- H Ehlken
- 1] Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany [2] University Medical Center Hamburg-Eppendorf, Hamburg D-20246, Germany
| | - S Krishna-Subramanian
- Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany
| | - L Ochoa-Callejero
- 1] Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany [2] Oncology Area, Center for Biomedical Research of La Rioja (CIBIR), 26006 Logroño, Spain
| | - V Kondylis
- Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany
| | - N E Nadi
- Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany
| | - B K Straub
- Institute of Pathology, University Hospital Heidelberg, INF 224, 69120 Heidelberg, Germany
| | - P Schirmacher
- Institute of Pathology, University Hospital Heidelberg, INF 224, 69120 Heidelberg, Germany
| | - H Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, 72 Huntley Street, WC1E 6BT London, UK
| | - G Kollias
- B.S.R.C. Alexander Fleming, 34 Fleming Street, Vari 16672, Greece
| | - M Pasparakis
- Institute for Genetics, Center for Molecular Medicine (CMMC), Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Strasse 26, Cologne D-50931, Germany
| |
Collapse
|
37
|
Abstract
The NF-κB family of inducible transcription factors is activated in response to a variety of stimuli. Amongst the best-characterized inducers of NF-κB are members of the TNF family of cytokines. Research on NF-κB and TNF have been tightly intertwined for more than 25 years. Perhaps the most compelling examples of the interconnectedness of NF-κB and the TNF have come from analysis of knock-out mice that are unable to activate NF-κB. Such mice die embryonically, however, deletion of TNF or TNFR1 can rescue the lethality thereby illustrating the important role of NF-κB as the key regulator of transcriptional responses to TNF. The physiological connections between NF-κB and TNF cytokines are numerous and best explored in articles focusing on a single TNF family member. Instead, in this review, we explore general mechanisms of TNF cytokine signaling, with a focus on the upstream signaling events leading to activation of the so-called canonical and noncanonical NF-κB pathways by TNFR1 and CD40, respectively.
Collapse
Affiliation(s)
- Matthew S Hayden
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA; Department of Dermatology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| | - Sankar Ghosh
- Department of Microbiology and Immunology, Columbia University, College of Physicians & Surgeons, New York, NY 10032, USA.
| |
Collapse
|
38
|
Dibra D, Mishra L, Li S. Molecular mechanisms of oncogene-induced inflammation and inflammation-sustained oncogene activation in gastrointestinal tumors: an under-appreciated symbiotic relationship. Biochim Biophys Acta Rev Cancer 2014; 1846:152-60. [PMID: 24821201 DOI: 10.1016/j.bbcan.2014.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 12/23/2022]
Abstract
Inflammation plays an integral part in tumor initiation. Specifically, patients with colitis, pancreatitis, or hepatitis have an increased susceptibility to cancer. The activation, mutation, and overexpression of oncogenes have been well documented in cell proliferation and transformation. Recently, oncogenes were found to also regulate the inflammatory milieu in tumors. Similarly, the inflammatory milieu can promote oncogene activation. In this review, we summarize advances of the symbiotic relationship oncogene activation and inflammation in gastrointestinal tumors such as colorectal, hepatic, and pancreatic tumors. NF-κB and STAT3 are the two most common pathways that are deregulated via these oncogenes. Understanding these interactions may yield effective therapeutic strategies for tumor prevention and treatment.
Collapse
Affiliation(s)
- Denada Dibra
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Lopa Mishra
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA
| | - Shulin Li
- Department of Pediatrics Research, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA.
| |
Collapse
|
39
|
Kung G, Dai P, Deng L, Kitsis RN. A novel role for the apoptosis inhibitor ARC in suppressing TNFα-induced regulated necrosis. Cell Death Differ 2014; 21:634-44. [PMID: 24440909 PMCID: PMC3950326 DOI: 10.1038/cdd.2013.195] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Revised: 11/15/2013] [Accepted: 11/25/2013] [Indexed: 12/15/2022] Open
Abstract
TNFα signaling can promote apoptosis or a regulated form of necrosis. ARC (apoptosis repressor with CARD (caspase recruitment domain)) is an endogenous inhibitor of apoptosis that antagonizes both the extrinsic (death receptor) and intrinsic (mitochondrial/ER) apoptosis pathways. We discovered that ARC blocks not only apoptosis but also necrosis. TNFα-induced necrosis was abrogated by overexpression of wild-type ARC but not by a CARD mutant that is also defective for inhibition of apoptosis. Conversely, knockdown of ARC exacerbated TNFα-induced necrosis, an effect that was rescued by reconstitution with wild-type, but not CARD-defective, ARC. Similarly, depletion of ARC in vivo exacerbated necrosis caused by infection with vaccinia virus, which elicits severe tissue damage through this pathway, and sensitized mice to TNFα-induced systemic inflammatory response syndrome. The mechanism underlying these effects is an interaction of ARC with TNF receptor 1 that interferes with recruitment of RIP1, a critical mediator of TNFα-induced regulated necrosis. These findings extend the role of ARC from an apoptosis inhibitor to a regulator of the TNFα pathway and an inhibitor of TNFα-mediated regulated necrosis.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis Regulatory Proteins/antagonists & inhibitors
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Cell Line, Tumor
- Fas-Associated Death Domain Protein/metabolism
- HMGB1 Protein/metabolism
- Humans
- MCF-7 Cells
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle Proteins/antagonists & inhibitors
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- NF-kappa B/metabolism
- Necrosis/chemically induced
- Necrosis/metabolism
- Necrosis/pathology
- Protein Binding
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- G Kung
- Departments of Cell Biology and Medicine, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - P Dai
- Dermatology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - L Deng
- Dermatology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - R N Kitsis
- Departments of Cell Biology and Medicine, Wilf Family Cardiovascular Research Institute, Albert Einstein Cancer Center, and Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
40
|
Baeck C, Tacke F. Balance of inflammatory pathways and interplay of immune cells in the liver during homeostasis and injury. EXCLI JOURNAL 2014; 13:67-81. [PMID: 26417243 PMCID: PMC4464465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 01/08/2014] [Indexed: 11/09/2022]
Abstract
Multiple potentially harmful stimuli challenge the liver, the chief metabolic and detoxifying organ of the human body. Due to its central anatomical location, continuous blood flow from the gastrointestinal tract through the hepatic sinusoids allows the metabolically active hepatocytes, the non-parenchymal cells and the various immune cell populations residing and patrolling in the liver to interact with antigens and microbiological components coming from the intestine. Cytokines are key mediators within the complex interplay of intrahepatic immune cells and hepatocytes, because they can activate effector functions of immune cells as well as hepatocytic intracellular signaling pathways controlling cellular homeostasis. Kupffer cells and liver-infiltrating monocyte-derived macrophages are primary sources of cytokines such as tumor necrosis factor (TNF). The liver is also enriched in natural killer (NK) and natural killer T (NKT) cells, which fulfill functions in pathogen defense, T cell recruitment and modulation of fibrogenic responses. TNF can activate specific intracellular pathways in hepatocytes that influence cell fate in different manners, e.g. pro-apoptotic signals via the caspase cascade, but also survival pathways, namely the nuclear factor (NF)-kappaB pathway. NF-kappaB regulates important functions in liver physiology and pathology. The exact dissection of the contribution of recruited and resident immune cells, their soluble cytokine and chemokine mediators and the intracellular hepatocytic response in liver homeostasis and injury could potentially identify novel targets for the treatment of acute and chronic liver disease, liver fibrosis or cirrhosis.
Collapse
Affiliation(s)
- Christer Baeck
- Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany,*To whom correspondence should be addressed: Frank Tacke, Department of Medicine III, RWTH-University Hospital Aachen, 52074 Aachen, Germany, Pauwelstraße 30; Phone: + 49-241-80-35848, Fax: + 49 -241-80-82455, E-mail:
| |
Collapse
|
41
|
Pharmacologic IKK/NF-κB inhibition causes antigen presenting cells to undergo TNFα dependent ROS-mediated programmed cell death. Sci Rep 2014; 4:3631. [PMID: 24406986 PMCID: PMC3887388 DOI: 10.1038/srep03631] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Accepted: 12/13/2013] [Indexed: 01/14/2023] Open
Abstract
Monocyte-derived antigen presenting cells (APC) are central mediators of the innate and adaptive immune response in inflammatory diseases. As such, APC are appropriate targets for therapeutic intervention to ameliorate certain diseases. APC differentiation, activation and functions are regulated by the NF-κB family of transcription factors. Herein, we examined the effect of NF-κB inhibition, via suppression of the IκB Kinase (IKK) complex, on APC function. Murine bone marrow-derived macrophages and dendritic cells (DC), as well as macrophage and DC lines, underwent rapid programmed cell death (PCD) after treatment with several IKK/NF-κB inhibitors through a TNFα-dependent mechanism. PCD was induced proximally by reactive oxygen species (ROS) formation, which causes a loss of mitochondrial membrane potential and activation of a caspase signaling cascade. NF-κB-inhibition-induced PCD of APC may be a key mechanism through which therapeutic targeting of NF-κB reduces inflammatory pathologies.
Collapse
|
42
|
NF-κB and cancer. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|
43
|
Cubero FJ, Singh A, Borkham-Kamphorst E, Nevzorova YA, Al Masaoudi M, Haas U, Boekschoten MV, Gassler N, Weiskirchen R, Muller M, Liedtke C, Trautwein C. TNFR1 determines progression of chronic liver injury in the IKKγ/Nemo genetic model. Cell Death Differ 2013; 20:1580-1592. [PMID: 23933814 PMCID: PMC3792433 DOI: 10.1038/cdd.2013.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 06/18/2013] [Accepted: 07/12/2013] [Indexed: 12/26/2022] Open
Abstract
Death receptor-mediated hepatocyte apoptosis is implicated in a wide range of liver diseases including viral and alcoholic hepatitis, ischemia/reperfusion injury, fulminant hepatic failure, cholestatic liver injury, as well as cancer. Deletion of NF-κB essential modulator in hepatocytes (IKKγ/Nemo) causes spontaneous progression of TNF-mediated chronic hepatitis to hepatocellular carcinoma (HCC). Thus, we analyzed the role of death receptors including TNFR1 and TRAIL in the regulation of cell death and the progression of liver injury in IKKγ/Nemo-deleted livers. We crossed hepatocyte-specific IKKγ/Nemo knockout mice (Nemo(Δhepa)) with constitutive TNFR1(-/-) and TRAIL(-/-) mice. Deletion of TNFR1, but not TRAIL, decreased apoptotic cell death, compensatory proliferation, liver fibrogenesis, infiltration of immune cells as well as pro-inflammatory cytokines, and indicators of tumor growth during the progression of chronic liver injury. These events were associated with diminished JNK activation. In contrast, deletion of TNFR1 in bone-marrow-derived cells promoted chronic liver injury. Our data demonstrate that TNF- and not TRAIL signaling determines the progression of IKKγ/Nemo-dependent chronic hepatitis. Additionally, we show that TNFR1 in hepatocytes and immune cells have different roles in chronic liver injury-a finding that has direct implications for treating chronic liver disease.
Collapse
Affiliation(s)
- F J Cubero
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - A Singh
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - E Borkham-Kamphorst
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - Y A Nevzorova
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - M Al Masaoudi
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - U Haas
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - M V Boekschoten
- Division of Human Nutrition, Metabolism and Genomics, Wageningen University, Wageningen, The Netherlands
| | - N Gassler
- Institute of Pathology, University Hospital, RWTH Aachen, Germany
| | - R Weiskirchen
- Institute of Clinical Chemistry and Pathobiochemistry, University Hospital, RWTH Aachen, Aachen, Germany
| | - M Muller
- Division of Human Nutrition, Metabolism and Genomics, Wageningen University, Wageningen, The Netherlands
| | - C Liedtke
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| | - C Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Aachen, Germany
| |
Collapse
|
44
|
Morris G, Berk M, Galecki P, Maes M. The emerging role of autoimmunity in myalgic encephalomyelitis/chronic fatigue syndrome (ME/cfs). Mol Neurobiol 2013; 49:741-56. [PMID: 24068616 DOI: 10.1007/s12035-013-8553-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 09/04/2013] [Indexed: 12/13/2022]
Abstract
The World Health Organization classifies myalgic encephalomyelitis/chronic fatigue syndrome (ME/cfs) as a nervous system disease. Together with other diseases under the G93 heading, ME/cfs shares a triad of abnormalities involving elevated oxidative and nitrosative stress (O&NS), activation of immuno-inflammatory pathways, and mitochondrial dysfunctions with depleted levels of adenosine triphosphate (ATP) synthesis. There is also abundant evidence that many patients with ME/cfs (up to around 60 %) may suffer from autoimmune responses. A wide range of reported abnormalities in ME/cfs are highly pertinent to the generation of autoimmunity. Here we review the potential sources of autoimmunity which are observed in people with ME/cfs. The increased levels of pro-inflammatory cytokines, e.g., interleukin-1 and tumor necrosis factor-α, and increased levels of nuclear factor-κB predispose to an autoimmune environment. Many cytokine abnormalities conspire to produce a predominance of effector B cells and autoreactive T cells. The common observation of reduced natural killer cell function in ME/cfs is a source of disrupted homeostasis and prolonged effector T cell survival. B cells may be pathogenic by playing a role in autoimmunity independent of their ability to produce antibodies. The chronic or recurrent viral infections seen in many patients with ME/cfs can induce autoimmunity by mechanisms involving molecular mimicry and bystander activation. Increased bacterial translocation, as observed in ME/cfs, is known to induce chronic inflammation and autoimmunity. Low ATP production and mitochondrial dysfunction is a source of autoimmunity by inhibiting apoptosis and stimulating necrotic cell death. Self-epitopes may be damaged by exposure to prolonged O&NS, altering their immunogenic profile and become a target for the host's immune system. Nitric oxide may induce many faces of autoimmunity stemming from elevated mitochondrial membrane hyperpolarization and blockade of the methionine cycle with subsequent hypomethylation of DNA. Here we also outline options for treatment involving rituximab and endotherapia.
Collapse
|
45
|
Shi X, Wang X, Li Q, Su M, Chew E, Wong ET, Lacza Z, Radda GK, Tergaonkar V, Han W. Nuclear factor κB (NF-κB) suppresses food intake and energy expenditure in mice by directly activating the Pomc promoter. Diabetologia 2013; 56:925-36. [PMID: 23370526 DOI: 10.1007/s00125-013-2831-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 12/28/2012] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS While chronic low-grade inflammation is associated with obesity, acute inflammation reduces food intake and leads to negative energy balance. Although both types of inflammation activate nuclear factor κB (NF-κB) signalling, it remains unclear how NF-κB activation results in opposite physiological responses in the two types of inflammation. The goal of this study was to address this question, and to understand the link between inflammation and leptin signalling. METHODS We studied the ability of NF-κB to modulate Pomc transcription, and how it impinges on signal transducer and activator of transcription 3 (STAT3)-mediated leptin signalling by using a combination of animal models, biochemical assays and molecular biology. RESULTS We report that suppression of food intake and physical movement with acute inflammation is not dependent on STAT3 activation in pro-opiomelanocortin (POMC) neurons. Under these conditions, activated NF-κB independently leads to increased Pomc transcription. Electrophoretic mobility shift assay and chromatin immunoprecipitation (ChIP) experiments reveal that NF-κB v-rel reticuloendotheliosis viral oncogene homologue A (avian) (RELA [also known as p65]) binds to the Pomc promoter region between -138 and -88 bp, which also harbours the trans-acting transcription factor 1 (SP1) binding site. We found significant changes in the methylation pattern at this region and reduced Pomc activation under chronic inflammation induced by a high-fat diet. Furthermore, RELA is unable to bind and activate transcription when the Pomc promoter is methylated. Finally, RELA binds to STAT3 and inhibits STAT3-mediated promoter activity, suggesting that RELA, possibly together with forkhead box-containing protein 1 (FOXO1), may prevent STAT3-mediated leptin activation of the Pomc promoter. CONCLUSIONS/INTERPRETATION Our study provides a mechanism for the involvement of RELA in the divergent regulation of energy homeostasis in acute and chronic inflammation.
Collapse
Affiliation(s)
- X Shi
- Singapore Bioimaging Consortium, Agency for Science, Technology and Research, #02-02 Helios, 11 Biopolis Way, Singapore 138667, Republic of Singapore
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Mouse Genetic Models Reveal Surprising Functions of IkB Kinase Alpha in Skin Development and Skin Carcinogenesis. Cancers (Basel) 2013; 5:170-83. [PMID: 24216703 PMCID: PMC3730312 DOI: 10.3390/cancers5010170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 01/25/2013] [Accepted: 02/06/2013] [Indexed: 01/05/2023] Open
Abstract
Gene knockout studies unexpectedly reveal a pivotal role for IκB kinase alpha (IKKα) in mouse embryonic skin development. Skin carcinogenesis experiments show that Ikkα heterozygous mice are highly susceptible to chemical carcinogen or ultraviolet B light (UVB) induced benign and malignant skin tumors in comparison to wild-type mice. IKKα deletion mediated by keratin 5 (K5).Cre or K15.Cre in keratinocytes induces epidermal hyperplasia and spontaneous skin squamous cell carcinomas (SCCs) in Ikkα floxed mice. On the other hand, transgenic mice overexpressing IKKα in the epidermis, under the control of a truncated loricrin promoter or K5 promoter, develop normal skin and show no defects in the formation of the epidermis and other epithelial organs, and the transgenic IKKα represses chemical carcinogen or UVB induced skin carcinogenesis. Moreover, IKKα deletion mediated by a mutation, which generates a stop codon in the Ikkα gene, has been reported in a human autosomal recessive lethal syndrome. Downregulated IKKα and Ikkα mutations and deletions are found in human skin SCCs. The collective evidence not only highlights the importance of IKKα in skin development, maintaining skin homeostasis, and preventing skin carcinogenesis, but also demonstrates that mouse models are extremely valuable tools for revealing the mechanisms underlying these biological events, leading our studies from bench side to bedside.
Collapse
|
47
|
Bandyopadhaya A, Kesarwani M, Que YA, He J, Padfield K, Tompkins R, Rahme LG. The quorum sensing volatile molecule 2-amino acetophenon modulates host immune responses in a manner that promotes life with unwanted guests. PLoS Pathog 2012; 8:e1003024. [PMID: 23166496 PMCID: PMC3499575 DOI: 10.1371/journal.ppat.1003024] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 09/26/2012] [Indexed: 12/25/2022] Open
Abstract
Increasing evidence indicates that bacterial quorum sensing (QS) signals are important mediators of immunomodulation. However, whether microbes utilize these immunomodulatory signals to maintain infection remain unclear. Here, we show that the Pseudomonas aeruginosa QS-regulated molecule 2-amino acetophenone (2-AA) modulates host immune responses in a manner that increases host ability to cope with this pathogen. Mice treated with 2-AA prior to infection had a 90% survival compared to 10% survival rate observed in the non-pretreated infected mice. Whilst 2-AA stimulation activates key innate immune response pathways involving mitogen-activated protein kinases (MAPKs), nuclear factor (NF)-κB, and pro-inflammatory cytokines, it attenuates immune response activation upon pretreatment, most likely by upregulating anti-inflammatory cytokines. 2-AA host pretreatment is characterized by a transcriptionally regulated block of c-JUN N-terminal kinase (JNK) and NF-κB activation, with relatively preserved activation of extracellular regulated kinase (ERK) 1/2. These kinase changes lead to CCAAT/enhancer-binding protein-β (c/EBPβ) activation and formation of the c/EBPβ-p65 complex that prevents NF-κB activation. 2-AA's aptitude for dampening the inflammatory processes while increasing host survival and pathogen persistence concurs with its ability to signal bacteria to switch to a chronic infection mode. Our results reveal a QS immunomodulatory signal that promotes original aspects of interkingdom communication. We propose that this communication facilitates pathogen persistence, while enabling host tolerance to infection.
Collapse
Affiliation(s)
- Arunava Bandyopadhaya
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Meenu Kesarwani
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Yok-Ai Que
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Jianxin He
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Katie Padfield
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Ronald Tompkins
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
| | - Laurence G. Rahme
- Department of Surgery, Harvard Medical School and Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, United States of America
- Shriners Hospitals for Children Boston, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
48
|
Roderburg C, Gautheron J, Luedde T. TNF-dependent signaling pathways in liver cancer: promising targets for therapeutic strategies? Dig Dis 2012; 30:500-7. [PMID: 23108306 DOI: 10.1159/000341700] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Liver cancer represents a growing health burden worldwide, and treatment options are still limited. Hepatocellular carcinoma (HCC), the most frequent primary hepatic malignancy, arises in most instances in chronically inflamed and fibrotic livers. However, current systemic molecular therapies against HCC are mainly focusing on tyrosine kinases involved in angiogenic and oncogenic signaling pathways, whereas the knowledge on the unique association between inflammation and carcinogenesis in the liver has not yet translated into preventive or therapeutic concepts against HCC. Tumor necrosis factor (TNF) is a cytokine derived from monocytes and various other immunological and parenchymal cells. Upon binding to its receptors, TNF activates different signaling cascades including the pro-apoptotic caspase cascade as well as inflammatory and stress-related pathways such as the NF-ĸB, p38MAPK, and Jun-(N)-terminal kinase (JNK) pathways. The role of TNF in cancer is controversial, since it was attributed both pro- and anti-carcinogenic functions. Its potential function in hepatocarcinogenesis has lately been investigated using genetically modified mouse models. These studies have highlighted that the various TNF-dependent signaling pathways withhold distinct functions in hepatocarcinogenesis, which are in part controversial and strongly depend on the experimental model system. Nevertheless, careful interpretation of findings in mouse models and critical consideration of their limitations might result in a new understanding of this complex pathway in hepatocarcinogenesis and thus might help identify the most promising targets in the TNF pathway and the appropriate clinical settings for future chemo-preventive or therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Christoph Roderburg
- Department of Internal Medicine III, University Hospital RWTH Aachen, Germany
| | | | | |
Collapse
|
49
|
Nakagawa H, Maeda S. Inflammation- and stress-related signaling pathways in hepatocarcinogenesis. World J Gastroenterol 2012; 18:4071-81. [PMID: 22919237 PMCID: PMC3422785 DOI: 10.3748/wjg.v18.i31.4071] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 05/28/2012] [Accepted: 06/08/2012] [Indexed: 02/06/2023] Open
Abstract
It has been established that cancer can be promoted and exacerbated by inflammation. Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide, and its long-term prognosis remains poor. Although HCC is a complex and heterogeneous tumor with several genomic mutations, it usually develops in the context of chronic liver damage and inflammation, suggesting that understanding the mechanism(s) of inflammation-mediated hepatocarcinogenesis is essential for the treatment and prevention of HCC. Chronic liver damage induces a persistent cycle of necro-inflammation and hepatocyte regeneration, resulting in genetic mutations in hepatocytes and expansion of initiated cells, eventually leading to HCC development. Recently, several inflammation- and stress-related signaling pathways have been identified as key players in these processes, which include the nuclear factor-κB, signal transducer and activator of transcription, and stress-activated mitogen- activated protein kinase pathways. Although these pathways may suggest potential therapeutic targets, they have a wide range of functions and complex crosstalk occurs among them. This review focuses on recent advances in our understanding of the roles of these signaling pathways in hepatocarcinogenesis.
Collapse
|
50
|
Affiliation(s)
- Sankar Ghosh
- Department of Microbiology & Immunology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | |
Collapse
|