1
|
Li Y, Song L, Yan X, Chi Y, Hu Y, Wang J, Robeldo D, Mukiibi R, Chen S. Orchestrated immune responses to Mycobacterium marinum natural infection in tongue sole (Cynoglossus semilaevis). FISH & SHELLFISH IMMUNOLOGY 2025; 158:110145. [PMID: 39837399 DOI: 10.1016/j.fsi.2025.110145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 01/23/2025]
Abstract
Mycobacterium marinum is a major pathogen in aquaculture, posing a substantial threat to the health and sustainability of tongue sole (Cynoglossus semilaevis) farming. This study investigated the genetic basis of immune response in tongue sole by comparing transcriptome profiles of liver and spleen tissues from symptomatic (susceptible) and healthy (resistant) individuals during a natural M. marinum outbreak. Transcriptomic analyses identified differentially expressed genes and enriched pathways related to immune responses. Key genes, including atp6ap1, gpi, and idh3a, were found to be crucial in immune response to M. marinum infection, involved in immune processes such as signal transduction, antigen processing, and metabolic pathways. Protein-protein interaction networks highlighted central hub genes such as nedd8, jun and junb, which play pivotal roles in immune regulation. These findings provide insights into the orchestrated immune responses to mycobacteriosis, which can inform selective breeding strategies for disease-resistant tongue sole strains. This is the first comprehensive transcriptome analysis of M. marinum natural infection in tongue sole, offering valuable data for future research and disease management in aquaculture.
Collapse
Affiliation(s)
- Yangzhen Li
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China
| | - Limin Song
- Tianjin Fisheries Research Institute, Tianjin, 300221, China
| | - Xu Yan
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Yong Chi
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Yuanri Hu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Jialin Wang
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China
| | - Diego Robeldo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, EH25 9RG, United Kingdom; Department of Genetics, Universidade de Santiago de Compostela, Santiago de Compostela, 15706, Spain
| | - Robert Mukiibi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, The University of Edinburgh, Midlothian, EH25 9RG, United Kingdom; Department of Animal Health, Behaviour and Welfare, Harper Adams University, Newport, Shropshire, TF10 8NB, United Kingdom.
| | - Songlin Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, 266071, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, Shandong, 266237, China.
| |
Collapse
|
2
|
Ishikawa C, Mori N. A New Strategy for Adult T-Cell Leukemia Treatment Targeting Glycogen Synthase Kinase-3β. Eur J Haematol 2024; 113:852-862. [PMID: 39239903 DOI: 10.1111/ejh.14300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/16/2024] [Accepted: 08/18/2024] [Indexed: 09/07/2024]
Abstract
OBJECTIVES The role of glycogen synthase kinase (GSK)-3β in adult T-cell leukemia (ATL) caused by human T-cell leukemia virus type 1 (HTLV-1) is paradoxical and enigmatic. Here, we investigated the role of GSK-3β and its potential as a therapeutic target for ATL. METHODS Cell proliferation/survival, cell cycle, apoptosis, and reactive oxygen species (ROS) generation were examined using the WST-8 assay, flow cytometry, and Hoechst 33342 staining, respectively. Expression of GSK-3β and cell cycle/death-related proteins, and survival signals was analyzed using RT-PCR, immunofluorescence staining, and immunoblotting. RESULTS HTLV-1-infected T-cell lines showed nuclear accumulation of GSK-3β. GSK-3β knockdown and its inhibition with 9-ING-41 and LY2090314 suppressed cell proliferation/survival. 9-ING-41 induced G2/M arrest by enhancing the expression of γH2AX, p53, p21, and p27, and suppressing the expression of CDK1, cyclin A/B, and c-Myc. It induced caspase-mediated apoptosis by decreasing the expression of Bcl-xL, Mcl-1, XIAP, c-IAP1/2, and survivin, and increasing the expression of Bak and Bax. 9-ING-41 also induced ferroptosis and necroptosis, promoted JNK phosphorylation, and suppressed IKKγ and JunB expression. It inhibited the phosphorylation of IκBα, Akt, and STAT3/5, induced ROS production, and reduced glycolysis-derived lactate levels. CONCLUSION GSK-3β functions as an oncogene in ATL and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
- Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Japan
| |
Collapse
|
3
|
Ishikawa C, Mori N. Inhibitory effect of a neddylation blockade on HTLV-1-infected T cells via modulation of NF-κB, AP-1, and Akt signaling. Leuk Lymphoma 2024; 65:978-988. [PMID: 38489672 DOI: 10.1080/10428194.2024.2328219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/03/2024] [Indexed: 03/17/2024]
Abstract
Adult T-cell leukemia (ATL), caused by HTLV-1, is the most lethal hematological malignancy. NEDD8-activating enzyme (NAE) is a component of the NEDD8 conjunction pathway that regulates cullin-RING ubiquitin ligase (CRL) activity. HTLV-1-infected T cells expressed higher levels of NAE catalytic subunit UBA3 than normal peripheral blood mononuclear cells. NAE1 knockdown inhibited proliferation of HTLV-1-infected T cells. The NAE1 inhibitor MLN4924 suppressed neddylation of cullin and inhibited the CRL-mediated turnover of tumor suppressor proteins. MLN4924 inhibited proliferation of HTLV-1-infected T cells by inducing DNA damage, leading to S phase arrest and caspase-dependent apoptosis. S phase arrest was associated with CDK2 and cyclin A downregulation. MLN4924-induced apoptosis was mediated by the upregulation of pro-apoptotic and downregulation of anti-apoptotic proteins. Furthermore, MLN4924 inhibited NF-κB, AP-1, and Akt signaling pathways and activated JNK. Therefore, neddylation inhibition is an attractive strategy for ATL therapy. Our findings support the use of MLN4924 in ATL clinical trials.
Collapse
Affiliation(s)
- Chie Ishikawa
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
- Division of Health Sciences, Transdisciplinary Research Organization for Subtropics and Island Studies, University of the Ryukyus, Nishihara, Okinawa, Japan
| | - Naoki Mori
- Department of Microbiology and Oncology, Graduate School of Medicine, University of the Ryukyus, Nishihara, Okinawa, Japan
| |
Collapse
|
4
|
Liu Z, Zhang J, Li S, Wang H, Ren B, Li J, Bao Z, Liu J, Guo M, Yang G, Chen L. Circadian control of ConA-induced acute liver injury and inflammatory response via Bmal1 regulation of Junb. JHEP Rep 2023; 5:100856. [PMID: 37791375 PMCID: PMC10542646 DOI: 10.1016/j.jhepr.2023.100856] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 10/05/2023] Open
Abstract
Background & Aims Circadian rhythms play significant roles in immune responses, and many inflammatory processes in liver diseases are associated with malfunctioning molecular clocks. However, the significance of the circadian clock in autoimmune hepatitis (AIH), which is characterised by immune-mediated hepatocyte destruction and extensive inflammatory cytokine production, remains unclear. Methods We tested the difference in susceptibility to the immune-mediated liver injury induced by concanavalin A (ConA) at various time points throughout a day in mice and analysed the effects of global, hepatocyte, or myeloid cell deletion of the core clock gene, Bmal1 (basic helix-loop-helix ARNT-like 1), on liver injury and inflammatory responses. Multiple molecular biology techniques and mice with macrophage-specific knockdown of Junb, a Bmal1 target gene, were used to investigate the involvement of Junb in the circadian control of ConA-induced hepatitis. Results The susceptibility to ConA-induced liver injury is highly dependent on the timing of ConA injection. The treatment at Zeitgeber time 0 (lights on) triggers the highest mortality as well as the severest liver injury and inflammatory responses. Further study revealed that this timing effect was driven by macrophage, but not hepatocyte, Bmal1. Mechanistically, Bmal1 controls the diurnal variation of ConA-induced hepatitis by directly regulating the circadian transcription of Junb and promoting M1 macrophage activation. Inhibition of Junb in macrophages blunts the administration time-dependent effect of ConA and attenuates liver injury. Moreover, we demonstrated that Junb promotes macrophage inflammation by regulating AKT and extracellular signal-regulated kinase (ERK) signalling pathways. Conclusions Our findings uncover a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced hepatitis and provide new insights into the prevention and treatment of AIH. Impact and Implications This study unveils a critical role of the Bmal1-Junb-AKT/ERK axis in the circadian control of ConA-induced liver injury, providing new insights into the prevention and treatment of immune-mediated hepatitis, including autoimmune hepatitis (AIH). The findings have scientific implications as they enhance our understanding of the circadian regulation of immune responses in liver diseases. Furthermore, clinically, this research offers opportunities for optimising treatment strategies in immune-mediated hepatitis by considering the timing of therapeutic interventions.
Collapse
Affiliation(s)
- Zhaiyi Liu
- School of Bioengineering, Dalian University of Technology, Dalian, China
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Jiayang Zhang
- Wuhu Hospital and Health Science Center, East China Normal University, Shanghai, China
| | - Shuyao Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hui Wang
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Baoyin Ren
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiazhi Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhiyue Bao
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Jiaxin Liu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Meina Guo
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guangrui Yang
- School of Clinical Medicine, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Lihong Chen
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
5
|
Ren FJ, Cai XY, Yao Y, Fang GY. JunB: a paradigm for Jun family in immune response and cancer. Front Cell Infect Microbiol 2023; 13:1222265. [PMID: 37731821 PMCID: PMC10507257 DOI: 10.3389/fcimb.2023.1222265] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Jun B proto-oncogene (JunB) is a crucial member of dimeric activator protein-1 (AP-1) complex, which plays a significant role in various physiological processes, such as placental formation, cardiovascular development, myelopoiesis, angiogenesis, endochondral ossification and epidermis tissue homeostasis. Additionally, it has been reported that JunB has great regulatory functions in innate and adaptive immune responses by regulating the differentiation and cytokine secretion of immune cells including T cells, dendritic cells and macrophages, while also facilitating the effector of neutrophils and natural killer cells. Furthermore, a growing body of studies have shown that JunB is involved in tumorigenesis through regulating cell proliferation, differentiation, senescence and metastasis, particularly affecting the tumor microenvironment through transcriptional promotion or suppression of oncogenes in tumor cells or immune cells. This review summarizes the physiological function of JunB, its immune regulatory function, and its contribution to tumorigenesis, especially focusing on its regulatory mechanisms within tumor-associated immune processes.
Collapse
Affiliation(s)
- Fu-jia Ren
- Department of Pharmacy, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| | - Xiao-yu Cai
- Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Yao
- Department of Pharmacy, Women’s Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Guo-ying Fang
- Department of Pharmacy, Hangzhou Women’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Griela E, Mountzouris KC. Nutrigenomic profiling of reduced specification diets and phytogenic inclusion effects on critical toll-like receptor signaling, mitogen-activated protein kinase-apoptosis, and PI3K-Akt-mTOR gene components along the broiler gut. Poult Sci 2023; 102:102675. [PMID: 37088046 PMCID: PMC10141502 DOI: 10.1016/j.psj.2023.102675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/23/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
The effects of concurrent reduction of dietary metabolizable energy (ME) and crude protein (CP) levels combined or not with the dietary inclusion of a phytogenic feed additive (PFA) were studied using a nutrigenomics approach. In particular, the expression of 26 critical genes relevant for inflammation control (TLR pathway), cellular apoptosis (MAPK pathway) cell growth and nutrient metabolism (PI3K-Akt-mTOR pathway) was profiled along the broiler intestine. Two dietary types (L and H) differing in metabolizable energy and crude protein levels (L: 95% and H: 100% of optimal Cobb 500 recommendations for ME and CP requirements) supplemented or not with PFA (- or +) and their interactions (L-, L+, H-, H+) were evaluated. There were only 3 total interactions (mTOR, IL8, and HRAS P < 0.05) between diet type and PFA inclusion indicating limited concurrent effects. Diet type, L upregulated genes related with inflammation mainly in the jejunum, ileum, and cecum (P < 0.05) and MAPK pathway in the ileum and cecum (P < 0.05). Moreover, diet type L negatively affected the expression of genes related to PI3K-Akt-mTOR pathway mainly in duodenum and cecum (P < 0.05). On the other hand, PFA inclusion downregulated (P < 0.05) genes related with TLR signaling pathway (TLR2B, MyD88, TLR3, IL8, LITAF) along the intestine and MAPK pathway genes (APO1, FOS) in jejunum (P < 0.05). Finally, PFA supplementation regulated nutrient sensing and metabolism in the cecum in a manner perceived as beneficial for growth. In conclusion, the study results highlight that the reduced ME and CP specifications, especially in the absence of PFA, regulate inflammation, apoptosis and nutrient metabolism processes at homeostatic control levels that hinder maximizing the availability of dietary energy and nutrients for growth purposes. Inclusion of PFA helped to adjust the respective homeostatic responses and control to levels supporting broiler performance, especially at reduced specification diets.
Collapse
Affiliation(s)
- Eirini Griela
- Laboratory of Nutritional Physiology and Feeding, Department of Animal Science, School of Animal Biosciences, Agricultural University of Athens, 11855 Athens, Greece
| | - Konstantinos C Mountzouris
- Laboratory of Nutritional Physiology and Feeding, Department of Animal Science, School of Animal Biosciences, Agricultural University of Athens, 11855 Athens, Greece.
| |
Collapse
|
7
|
Pérez-Benavente B, Fathinajafabadi A, de la Fuente L, Gandía C, Martínez-Férriz A, Pardo-Sánchez JM, Milián L, Conesa A, Romero OA, Carretero J, Matthiesen R, Jariel-Encontre I, Piechaczyk M, Farràs R. New roles for AP-1/JUNB in cell cycle control and tumorigenic cell invasion via regulation of cyclin E1 and TGF-β2. Genome Biol 2022; 23:252. [PMID: 36494864 PMCID: PMC9733061 DOI: 10.1186/s13059-022-02800-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 10/20/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND JUNB transcription factor contributes to the formation of the ubiquitous transcriptional complex AP-1 involved in the control of many physiological and disease-associated functions. The roles of JUNB in the control of cell division and tumorigenic processes are acknowledged but still unclear. RESULTS Here, we report the results of combined transcriptomic, genomic, and functional studies showing that JUNB promotes cell cycle progression via induction of cyclin E1 and repression of transforming growth factor (TGF)-β2 genes. We also show that high levels of JUNB switch the response of TGF-β2 stimulation from an antiproliferative to a pro-invasive one, induce endogenous TGF-β2 production by promoting TGF-β2 mRNA translation, and enhance tumor growth and metastasis in mice. Moreover, tumor genomic data indicate that JUNB amplification associates with poor prognosis in breast and ovarian cancer patients. CONCLUSIONS Our results reveal novel functions for JUNB in cell proliferation and tumor aggressiveness through regulation of cyclin E1 and TGF-β2 expression, which might be exploited for cancer prognosis and therapy.
Collapse
Affiliation(s)
| | | | - Lorena de la Fuente
- Centro de Investigación Príncipe Felipe, Valencia, Spain
- Present Address: PerkinElmer Informatics, Tres Cantos, Madrid, Spain
| | | | | | | | - Lara Milián
- Department of Pathology, Faculty of Medicine and Dentistry, Universitat de València, Valencia, Spain
- INCLIVA Biomedical Research Institute, 46010, Valencia, Spain
| | - Ana Conesa
- Spanish National Research Council, Institute for Integrative Systems Biology, Paterna, Valencia, Spain
- Microbiology and Cell Science, University of Florida, Gainesville, FL, USA
| | - Octavio A Romero
- Cancer Genetics Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Julián Carretero
- Departament de Fisiologia, Facultat de Farmacia, Universitat de València, Burjassot, Valencia, Spain
| | - Rune Matthiesen
- Computational and Experimental Biology Group, NOVA Medical School-Research, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Isabelle Jariel-Encontre
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
- Present address: IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France
| | - Rosa Farràs
- Centro de Investigación Príncipe Felipe, Valencia, Spain.
| |
Collapse
|
8
|
Hamway Y, Zimmermann K, Blommers MJJ, Sousa MV, Häberli C, Kulkarni S, Skalicky S, Hackl M, Götte M, Keiser J, da Costa CP, Spangenberg T, Azzaoui K. Modulation of Host-Parasite Interactions with Small Molecules Targeting Schistosoma mansoni microRNAs. ACS Infect Dis 2022; 8:2028-2034. [PMID: 36098656 PMCID: PMC9578036 DOI: 10.1021/acsinfecdis.2c00360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Parasites use different strategies of communication with their hosts. One communication channel that has been studied in recent years is the use of vesicle microRNAs to influence the host immune system by trematodes. sma-microRNA-10, secreted from Schistosoma mansoni, has been shown to influence the fate of host T-cells through manipulation of the NF-κB pathway. We have identified low molecular weight tool compounds that can interfere with this microRNA-mediated manipulation of the host immune system. We used a fragment-based screening approach by means of nuclear magnetic resonance (NMR) to identify binders to the precursor of the parasite sma-microRNA-10 present in their extracellular vesicles. The small fragments identified were used to select larger molecules. These molecules were shown to counteract the inhibition of NF-κB activity by sma-microRNA-10 in cell-based assays.
Collapse
Affiliation(s)
- Youssef Hamway
- Institute
for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675Munich, Germany,Center
for Global Health, TUM School of Medicine, Technical University of Munich, 81675Munich, Germany
| | - Kaspar Zimmermann
- Saverna
Therapeutics AG, Gewerbestrasse
24, 4123Allschwil, Switzerland
| | | | - Mariana V. Sousa
- Institute
for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675Munich, Germany,Center
for Global Health, TUM School of Medicine, Technical University of Munich, 81675Munich, Germany
| | - Cécile Häberli
- Department
of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123Allschwil, Switzerland,University
of Basel, Petersplatz
1, 4001Basel, Switzerland
| | - Shashank Kulkarni
- EMD
Serono Research & Development Institute, Inc. (a Business of Merck KGaA, Darmstadt, Germany), Billerica, Massachusetts01821, United States
| | | | | | - Marjo Götte
- Saverna
Therapeutics AG, Gewerbestrasse
24, 4123Allschwil, Switzerland
| | - Jennifer Keiser
- Department
of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123Allschwil, Switzerland,University
of Basel, Petersplatz
1, 4001Basel, Switzerland
| | - Clarissa Prazeres da Costa
- Institute
for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, 81675Munich, Germany,Center
for Global Health, TUM School of Medicine, Technical University of Munich, 81675Munich, Germany
| | - Thomas Spangenberg
- Global
Health Institute of Merck, Ares Trading S.A., a subsidiary of Merck KGaA, Darmstadt, Route de
Crassier 1, 1262Eysins, Switzerland,
| | - Kamal Azzaoui
- Saverna
Therapeutics AG, Gewerbestrasse
24, 4123Allschwil, Switzerland,
| |
Collapse
|
9
|
The Early Immune Response of Lymphoid and Myeloid Head-Kidney Cells of Rainbow Trout (Oncorhynchus mykiss) Stimulated with Aeromonas salmonicida. FISHES 2022. [DOI: 10.3390/fishes7010012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The teleost head kidney is a highly relevant immune organ, and myeloid cells play a major role in this organ’s innate and adaptive immune responses. Because of their complexity, the early phases of the innate immune reaction of fish against bacteria are still poorly understood. In this study, naïve rainbow trout were stimulated with inactivated A. salmonicida and sampled at 12 h, 24 h and 7 d poststimulation. Cells from the head kidney were magnetically sorted with a monoclonal antibody mAB21 to obtain one (MAb21-positive) fraction enriched with myeloid cells and one (MAb21-negative) fraction enriched with lymphocytes and thrombocytes. The gene expression pattern of the resulting cell subpopulations was analysed using a panel of 43 immune-related genes. The results show an overall downregulation of the complement pathway and cytokine production at the considered time points. Some of the selected genes may be considered as parameters for diagnosing bacterial furunculosis of rainbow trout.
Collapse
|
10
|
IκBα is required for full transcriptional induction of some NFκB-regulated genes in response to TNF in MCF-7 cells. NPJ Syst Biol Appl 2021; 7:42. [PMID: 34853340 PMCID: PMC8636565 DOI: 10.1038/s41540-021-00204-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory stimuli triggers the degradation of three inhibitory κB (IκB) proteins, allowing for nuclear translocation of nuclear factor-κB (NFκB) for transcriptional induction of its target genes. Of these three, IκBα is a well-known negative feedback regulator that limits the duration of NFκB activity. We sought to determine whether IκBα's role in enabling or limiting NFκB activation is important for tumor necrosis factor (TNF)-induced gene expression in human breast cancer cells (MCF-7). Contrary to our expectations, many more TNF-response genes showed reduced induction than enhanced induction in IκBα knockdown cells. Mathematical modeling was used to investigate the underlying mechanism. We found that the reduced activation of some NFκB target genes in IκBα-deficient cells could be explained by the incoherent feedforward loop (IFFL) model. In addition, for a subset of genes, prolonged NFκB activity due to loss of negative feedback control did not prolong their transient activation; this implied a multi-state transcription cycle control of gene induction. Genes encoding key inflammation-related transcription factors, such as JUNB and KLF10, were found to be best represented by a model that contained both the IFFL and the transcription cycle motif. Our analysis sheds light on the regulatory strategies that safeguard inflammatory gene expression from overproduction and repositions the function of IκBα not only as a negative feedback regulator of NFκB but also as an enabler of NFκB-regulated stimulus-responsive inflammatory gene expression. This study indicates the complex involvement of IκBα in the inflammatory response to TNF that is induced by radiation therapy in breast cancer.
Collapse
|
11
|
Giordano R, Petersen KK, Andersen HH, Lichota J, Valeriani M, Simonsen O, Arendt-Nielsen L. Preoperative serum circulating microRNAs as potential biomarkers for chronic postoperative pain after total knee replacement. Mol Pain 2021; 16:1744806920962925. [PMID: 33021154 PMCID: PMC7543153 DOI: 10.1177/1744806920962925] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Chronic postoperative pain affects approximately 20% of patients with knee
osteoarthritis after total knee replacement. Circulating microRNAs can be
found in serum and might act as biomarkers in a variety of diseases. The
current study aimed to investigate the preoperative expression of
circulating microRNAs as potential predictive biomarkers for the development
of chronic postoperative pain in the year following total knee
replacement. Methods Serum samples, collected preoperatively from 136 knee osteoarthritis
patients, were analyzed for 21 circulatory microRNAs. Pain intensity was
assessed using a visual analog scale before and one year after total knee
replacement. Patients were divided into a low-pain relief group (pain relief
percentage <30%) and a high-pain relief group (pain relief percentage
>30%) based on their pain relief one year after total knee replacement,
and differences in microRNAs expression were analyzed between the two
groups. Results We found that three microRNAs were preoperatively dysregulated in serum in
the low-pain relief group compared with the high-pain relief group.
MicroRNAs hsa-miR-146a-5p, -145-5p, and -130 b-3p exhibited fold changes of
1.50, 1.55, and 1.61, respectively, between the groups (all P
values < 0.05). Hsa-miR-146a-5p and preoperative pain intensity
correlated positively with postoperative pain relief (respectively,
R = 0.300, P = 0.006; R = 0.500, P < 0.001). Discussion This study showed that patients with a low postoperative pain relief present
a dysregulation of circulating microRNAs. Altered circulatory microRNAs
expression correlated with postoperative pain relief, indicating that
microRNAs can serve as predictive biomarkers of pain outcome after surgery
and hence may foster new strategies for preventing chronic postoperative
pain after total knee replacement (TKR).
Collapse
Affiliation(s)
- Rocco Giordano
- Center for Neuroplasticity and Pain, SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Kristian Kjær Petersen
- Center for Neuroplasticity and Pain, SMI, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark.,Center for Sensory-Motor Interaction, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Hjalte Holm Andersen
- Center for Sensory-Motor Interaction, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Jacek Lichota
- Laboratory of Metabolism Modifying Medicine, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| | - Massimiliano Valeriani
- Center for Sensory-Motor Interaction, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark.,Child Neurology Unit, Department of Neuroscience and Neurorehabilitation, Headache Center, Bambino Gesù Children's Hospital, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Ole Simonsen
- Orthopedic Surgery Research Unit, Aalborg University Hospital, Aalborg, Denmark
| | - Lars Arendt-Nielsen
- Center for Sensory-Motor Interaction, Department of Health Science and Technology, Faculty of Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
12
|
Harman JL, Dobnikar L, Chappell J, Stokell BG, Dalby A, Foote K, Finigan A, Freire-Pritchett P, Taylor AL, Worssam MD, Madsen RR, Loche E, Uryga A, Bennett MR, Jørgensen HF. Epigenetic Regulation of Vascular Smooth Muscle Cells by Histone H3 Lysine 9 Dimethylation Attenuates Target Gene-Induction by Inflammatory Signaling. Arterioscler Thromb Vasc Biol 2019; 39:2289-2302. [PMID: 31434493 PMCID: PMC6818986 DOI: 10.1161/atvbaha.119.312765] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Vascular inflammation underlies cardiovascular disease. Vascular smooth muscle cells (VSMCs) upregulate selective genes, including MMPs (matrix metalloproteinases) and proinflammatory cytokines upon local inflammation, which directly contribute to vascular disease and adverse clinical outcome. Identification of factors controlling VSMC responses to inflammation is therefore of considerable therapeutic importance. Here, we determine the role of Histone H3 lysine 9 di-methylation (H3K9me2), a repressive epigenetic mark that is reduced in atherosclerotic lesions, in regulating the VSMC inflammatory response. Approach and Results: We used VSMC-lineage tracing to reveal reduced H3K9me2 levels in VSMCs of arteries after injury and in atherosclerotic lesions compared with control vessels. Intriguingly, chromatin immunoprecipitation showed H3K9me2 enrichment at a subset of inflammation-responsive gene promoters, including MMP3, MMP9, MMP12, and IL6, in mouse and human VSMCs. Inhibition of G9A/GLP (G9A-like protein), the primary enzymes responsible for H3K9me2, significantly potentiated inflammation-induced gene induction in vitro and in vivo without altering NFκB (nuclear factor kappa-light-chain-enhancer of activated B cell) and MAPK (mitogen-activated protein kinase) signaling. Rather, reduced G9A/GLP activity enhanced inflammation-induced binding of transcription factors NFκB-p65 and cJUN to H3K9me2 target gene promoters MMP3 and IL6. Taken together, these results suggest that promoter-associated H3K9me2 directly attenuates the induction of target genes in response to inflammation in human VSMCs. CONCLUSIONS This study implicates H3K9me2 in regulating the proinflammatory VSMC phenotype. Our findings suggest that reduced H3K9me2 in disease enhance binding of NFκB and AP-1 (activator protein-1) transcription factors at specific inflammation-responsive genes to augment proinflammatory stimuli in VSMC. Therefore, H3K9me2-regulation could be targeted clinically to limit expression of MMPs and IL6, which are induced in vascular disease.
Collapse
Affiliation(s)
- Jennifer L. Harman
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Lina Dobnikar
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
- Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom (L.D., P.F.-P.)
| | - Joel Chappell
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Benjamin G. Stokell
- Statistical Laboratory, Centre for Mathematical Sciences, University of Cambridge, United Kingdom (B.G.S.)
| | - Amanda Dalby
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Kirsty Foote
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Alison Finigan
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | | | - Annabel L. Taylor
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Matthew D. Worssam
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Ralitsa R. Madsen
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Elena Loche
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Anna Uryga
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Martin R. Bennett
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| | - Helle F. Jørgensen
- From the Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (J.L.H., L.D., J.C., A.D., K.F., A.F., A.L.T., M.D.W., R.R.M., E.L., A.U., M.R.B., H.F.J.)
| |
Collapse
|
13
|
Safflower Yellow B Protects Brain against Cerebral Ischemia Reperfusion Injury through AMPK/NF-kB Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:7219740. [PMID: 30854014 PMCID: PMC6378026 DOI: 10.1155/2019/7219740] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 01/15/2019] [Indexed: 11/24/2022]
Abstract
Inflammation had showed its important role in the pathogenesis of cerebral ischemia and secondary damage. Safflower yellow B (SYB) had neuroprotective effects against oxidative stress-induced brain injuries, but the mechanisms were still largely unknown to us. In this study, we tried to investigate the anti-inflammation effects of SYB and the possible roles of AMPK/NF-κB signaling pathway on these protective effects. In vivo, brain ischemia/reperfusion (I/R) was induced by transient middle cerebral artery occlusion for 2 h and reperfusion for 20 h. Neurofunctional evaluation, infarction area, and brain water contents were measured. Brain injury markers and inflammatory cytokines levels were measured by ELISA kits. In vitro, cell viability, apoptosis, and LDH leakage were measured after I/R in PC12 cells. The expression and phosphorylation levels of AMPK, NF-κB p65, and P-IκB-α in cytoplasm and nuclear were measured by Western blotting. SiRNA experiment was performed to certify the role of AMPK. The results showed SYB reduced infarct size, improved neurological outcomes, and inhibited brain injury after I/R. In vitro test, SYB treatment alleviated PC12 cells injury and apoptosis and inhibited the inflammatory cytokines (IL-1, IL-6, TNF-α, and COX-2) in a dose-dependent manner. SYB treatment induced AMPK phosphorylation and inhibited NF-κB p65 nuclear translocation both in brain and in PC12 cells. Further studies also showed that the inhibition of NF-κB activity of SYB was through AMPK. In conclusion, SYB protected brain I/R injury through reducing expression of inflammatory cytokines and this effect might be partly due to the inhibition of NF-κB mediated by AMPK.
Collapse
|
14
|
Human Intervention Study to Assess the Effects of Supplementation with Olive Leaf Extract on Peripheral Blood Mononuclear Cell Gene Expression. Int J Mol Sci 2016; 17:ijms17122019. [PMID: 27918443 PMCID: PMC5187819 DOI: 10.3390/ijms17122019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 11/21/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022] Open
Abstract
Olive leaf extract (OLE) has been used for many years for its putative health benefits, but, to date, scientific evidence for the basis of these effects has been weak. Although recent literature has described a link between ailments such as cardiovascular disease, diabetes and cancer and a protective effect of polyphenols in the OLE, the mode of action is still unclear. Here, we describe a double-blinded placebo (PBO)-controlled trial, in which gene expression profiles of peripheral blood mononuclear cells from healthy male volunteers (n = 29) were analysed to identify genes that responded to OLE, following an eight-week intervention with 20 mL daily consumption of either OLE or PBO. Differences between groups were determined using an adjusted linear model. Subsequent analyses indicated downregulation of genes important in inflammatory pathways, lipid metabolism and cancer as a result of OLE consumption. Gene expression was verified by real-time PCR for three genes (EGR1, COX-2 and ID3). The results presented here suggest that OLE consumption may result in health benefits through influencing the expression of genes in inflammatory and metabolic pathways. Future studies with a larger study group, including male and female participants, looking into direct effects of OLE on lipid metabolism and inflammation are warranted.
Collapse
|
15
|
Chouvardas P, Kollias G, Nikolaou C. Inferring active regulatory networks from gene expression data using a combination of prior knowledge and enrichment analysis. BMC Bioinformatics 2016; 17 Suppl 5:181. [PMID: 27295045 PMCID: PMC4905609 DOI: 10.1186/s12859-016-1040-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Background Under both physiological and pathological conditions gene expression programs are shaped through the interplay of regulatory proteins and their gene targets, interactions between which form intricate gene regulatory networks (GRN). While the assessment of genome-wide expression for the complete set of genes at a given condition has become rather straight-forward and is performed routinely, we are still far from being able to infer the topology of gene regulation simply by analyzing its “descendant” expression profile. In this work we are trying to overcome the existing limitations for the inference and study of such regulatory networks. We are combining our approach with state-of-the-art gene set enrichment analyses in order to create a tool, called Regulatory Network Enrichment Analysis (RNEA) that will prioritize regulatory and functional characteristics of a genome-wide expression experiment. Results RNEA combines prior knowledge, originating from manual literature curation and small-scale experimental data, to construct a reference network of interactions and then uses enrichment analysis coupled with a two-level hierarchical parsing of the network, to infer the most relevant subnetwork for a given experimental setting. It is implemented as an R package, currently supporting human and mouse datasets and was herein tested on one test case for each of the two organisms. In both cases, RNEA’s gene set enrichment analysis was comparable to state-of-the-art methodologies. Moreover, through its distinguishing feature of regulatory subnetwork reconstruction, RNEA was able to define the key transcriptional regulators for the studied systems as supported from the literature. Conclusions RNEA constitutes a novel computational approach to obtain regulatory interactions directly from a genome-wide expression profile. Its simple implementation, with minimal requirements from the user is coupled with easy-to-parse enrichment lists and a subnetwork file that may be readily visualized to reveal the most important components of the regulatory hierarchy. The combination of prior information and novel concept of a hierarchical reconstruction of regulatory interactions makes RNEA a very useful tool for a first-level interpretation of gene expression profiles. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1040-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Panagiotis Chouvardas
- Biomedical Sciences Research Center "Alexander Fleming", Vari, 16672, Greece.,Department of Physiology, Medical School, University of Athens, Athens, 11527, Greece
| | - George Kollias
- Division of Immunology, Biomedical Sciences Research Center "Alexander Fleming", Vari, 16672, Greece.,Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, 11527, Greece
| | - Christoforos Nikolaou
- Biomedical Sciences Research Center "Alexander Fleming", Vari, 16672, Greece. .,Computational Genomics Group, Department of Biology, University of Crete, Voutes Campus, Heraklion, 70013, Greece.
| |
Collapse
|
16
|
Cahill CM, Zhu W, Oziolor E, Yang YJ, Tam B, Rajanala S, Rogers JT, Walker WA. Differential Expression of the Activator Protein 1 Transcription Factor Regulates Interleukin-1ß Induction of Interleukin 6 in the Developing Enterocyte. PLoS One 2016; 11:e0145184. [PMID: 26799482 PMCID: PMC4723075 DOI: 10.1371/journal.pone.0145184] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/30/2015] [Indexed: 01/05/2023] Open
Abstract
The innate immune response is characterized by activation of transcription factors, nuclear factor kappa B and activator protein-1 and their downstream targets, the pro-inflammatory cytokines including interleukin 1β and interleukin 6. Normal development of this response in the intestine is critical to survival of the human neonate and delays can cause the onset of devastating inflammatory diseases such as necrotizing enterocolitis. Previous studies have addressed the role of nuclear factor kappa B in the development of the innate immune response in the enterocyte, however despite its central role in the control of multiple pro-inflammatory cytokine genes, little is known on the role of Activator Protein 1 in this response in the enterocyte. Here we show that the canonical Activator Protein 1 members, cJun and cFos and their upstream kinases JNK and p38 play an essential role in the regulation of interleukin 6 in the immature enterocyte. Our data supports a model whereby the cFos/cJun heterodimer and the more potent cJun homodimer downstream of JNK are replaced by less efficient JunD containing dimers, contributing to the decreased responsiveness to interleukin 1β and decreased interleukin 6 secretion observed in the mature enterocyte. The tissue specific expression of JunB in colonocytes and colon derived tissues together with its ability to repress Interleukin-1β induction of an Interleukin-6 gene reporter in the NCM-460 colonocyte suggests that induction of JunB containing dimers may offer an attractive therapeutic strategy for the control of IL-6 secretion during inflammatory episodes in this area of the intestine
Collapse
Affiliation(s)
- Catherine M. Cahill
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Charlestown, Massachusetts, United States of America
- * E-mail:
| | - Weishu Zhu
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital for Children, and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Elias Oziolor
- Department. of Environmental Science, Baylor University One Bear Place #97266, Waco, Texas, United States of America
| | - Yao-Jong Yang
- Departments of Pediatrics and Internal Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bosco Tam
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Susruthi Rajanala
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital, and Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - W. Allan Walker
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital for Children, and Harvard Medical School, Charlestown, Massachusetts, United States of America
| |
Collapse
|
17
|
Fontana MF, Baccarella A, Pancholi N, Pufall MA, Herbert DR, Kim CC. JUNB is a key transcriptional modulator of macrophage activation. THE JOURNAL OF IMMUNOLOGY 2014; 194:177-86. [PMID: 25472994 DOI: 10.4049/jimmunol.1401595] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Activated macrophages are crucial for restriction of microbial infection but may also promote inflammatory pathology in a wide range of both infectious and sterile conditions. The pathways that regulate macrophage activation are therefore of great interest. Recent studies in silico have putatively identified key transcription factors that may control macrophage activation, but experimental validation is lacking. In this study, we generated a macrophage regulatory network from publicly available microarray data, employing steps to enrich for physiologically relevant interactions. Our analysis predicted a novel relationship between the AP-1 family transcription factor Junb and the gene Il1b, encoding the pyrogen IL-1β, which macrophages express upon activation by inflammatory stimuli. Previously, Junb has been characterized primarily as a negative regulator of the cell cycle, whereas AP-1 activity in myeloid inflammatory responses has largely been attributed to c-Jun. We confirmed experimentally that Junb is required for full expression of Il1b, and of additional genes involved in classical inflammation, in macrophages treated with LPS and other immunostimulatory molecules. Furthermore, Junb modulates expression of canonical markers of alternative activation in macrophages treated with IL-4. Our results demonstrate that JUNB is a significant modulator of both classical and alternative macrophage activation. Further, this finding provides experimental validation for our network modeling approach, which will facilitate the future use of gene expression data from open databases to reveal novel, physiologically relevant regulatory relationships.
Collapse
Affiliation(s)
- Mary F Fontana
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| | - Alyssa Baccarella
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| | - Nidhi Pancholi
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| | - Miles A Pufall
- Department of Biochemistry, University of Iowa, Iowa City, IA 52242
| | - De'Broski R Herbert
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| | - Charles C Kim
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143; and
| |
Collapse
|
18
|
Moquet-Torcy G, Tolza C, Piechaczyk M, Jariel-Encontre I. Transcriptional complexity and roles of Fra-1/AP-1 at the uPA/Plau locus in aggressive breast cancer. Nucleic Acids Res 2014; 42:11011-24. [PMID: 25200076 PMCID: PMC4176185 DOI: 10.1093/nar/gku814] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Plau codes for the urokinase-type plasminogen activator (uPA), critical in cancer metastasis. While the mechanisms driving its overexpression in tumorigenic processes are unknown, it is regulated by the AP-1 transcriptional complex in diverse situations. The AP-1 component Fra-1 being overexpressed in aggressive breast cancers, we have addressed its role in the overexpression of Plau in the highly metastatic breast cancer model cell line MDA-MB231 using ChIP, pharmacological and RNAi approaches. Plau transcription appears controlled by 2 AP-1 enhancers located -1.9 (ABR-1.9) and -4.1 kb (ABR-4.1) upstream of the transcription start site (TSS) of the uPA-coding mRNA, Plau-001, that bind Fra-1. Surprisingly, RNA Pol II is not recruited only at the Plau-001 TSS but also upstream in the ABR-1.9 and ABR-4.1 region. Most Pol II molecules transcribe short and unstable RNAs while tracking down toward the TSS, where there are converted into Plau-001 mRNA-productive species. Moreover, a minority of Pol II molecules transcribes a low abundance mRNA of unknown function called Plau-004 from the ABR-1.9 domain, whose expression is tempered by Fra-1. Thus, we unveil a heretofore-unsuspected transcriptional complexity at Plau in a reference metastatic breast cancer cell line with pleiotropic effects for Fra-1, providing novel information on AP-1 transcriptional action.
Collapse
Affiliation(s)
- Gabriel Moquet-Torcy
- Institut de Génétique Moléculaire de Montpellier UMR 5535, CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5, France Université Montpellier 1, 5 Bd Henry IV, 34967 Montpellier cedex 2, France
| | - Claire Tolza
- Institut de Génétique Moléculaire de Montpellier UMR 5535, CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5, France Université Montpellier 1, 5 Bd Henry IV, 34967 Montpellier cedex 2, France
| | - Marc Piechaczyk
- Institut de Génétique Moléculaire de Montpellier UMR 5535, CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5, France Université Montpellier 1, 5 Bd Henry IV, 34967 Montpellier cedex 2, France
| | - Isabelle Jariel-Encontre
- Institut de Génétique Moléculaire de Montpellier UMR 5535, CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5, France Université Montpellier 1, 5 Bd Henry IV, 34967 Montpellier cedex 2, France
| |
Collapse
|
19
|
Nuzzo AM, Giuffrida D, Zenerino C, Piazzese A, Olearo E, Todros T, Rolfo A. JunB/cyclin-D1 imbalance in placental mesenchymal stromal cells derived from preeclamptic pregnancies with fetal-placental compromise. Placenta 2014; 35:483-90. [PMID: 24780198 DOI: 10.1016/j.placenta.2014.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 04/03/2014] [Accepted: 04/07/2014] [Indexed: 11/19/2022]
Abstract
INTRODUCTION In the present study, we characterized the expression of Activating Protein 1 (AP-1) factors, key cell cycle regulators, in primary placental mesenchymal stromal cells (PDMSCs) derived from normal and preeclamptic (PE) pregnancies with fetal-placental compromise. METHODS PDMSCs were isolated from control (n = 20) and preeclamptic (n = 24) placentae. AP-1 expression was determined by semi-quantitative RT-PCR (sqRT-PCR), Real Time PCR and Western Blot assay. PDMSCs were plated and JunB siRNA was performed. JunB and Cyclin-D1 expression were assessed by Real Time and Western Blot analyses. RESULTS JunB expression was significantly increased while Cyclin-D1 expression was significantly down-regulated in PE relative to control PDMSCs. JunB siRNA was accompanied by JunB down-regulation and increased Cyclin-D1 in normal PDMSCs. CONCLUSIONS We described, for the first time, AP-1 expression in PDMSCs derived from physiological and PE placentae. Importantly, we demonstrated that JunB over-expression in PE-PDMSCs affects Cyclin-D1 regulation. Our data suggest a possible contribution of these pathological placental cells to the altered cell cycle regulation typical of preeclamptic placentae.
Collapse
Affiliation(s)
- A M Nuzzo
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - D Giuffrida
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - C Zenerino
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - A Piazzese
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - E Olearo
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - T Todros
- Dept. of Surgical Sciences, University of Turin, Turin, Italy
| | - A Rolfo
- Dept. of Surgical Sciences, University of Turin, Turin, Italy.
| |
Collapse
|
20
|
Pooley NJ, Tacchi L, Secombes CJ, Martin SAM. Inflammatory responses in primary muscle cell cultures in Atlantic salmon (Salmo salar). BMC Genomics 2013; 14:747. [PMID: 24180744 PMCID: PMC3819742 DOI: 10.1186/1471-2164-14-747] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 10/26/2013] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The relationship between fish health and muscle growth is critical for continued expansion of the aquaculture industry. The effect of immune stimulation on the expression of genes related to the energy balance of fish is poorly understood. In mammals immune stimulation results in major transcriptional changes in muscle, potentially to allow a reallocation of amino acids for use in the immune response and energy homeostasis. The aim of this study was to investigate the effects of immune stimulation on fish muscle gene expression. RESULTS Atlantic salmon (Salmo salar) primary muscle cell cultures were stimulated with recombinant (r)IL-1β, a major proinflammatory cytokine, for 24 h in order to simulate an acute immune response. The transcriptomic response was determined by RNA hybridization to a 4 × 44 K Agilent Atlantic salmon microarray platform. The rIL-1β stimulation induced the expression of genes related to both the innate and adaptive immune systems. In addition there were highly significant changes in the expression of genes related to regulation of the cell cycle, growth/structural proteins, proteolysis and lipid metabolism. Of interest were a number of IGF binding proteins that were differentially expressed, which may demonstrate cross talk between the growth and immune systems. CONCLUSION We show rIL-1β modulates the expression of not only immune related genes, but also that of genes involved in processes related to growth and metabolism. Co-stimulation of muscle cells with both rIGF-I and rIL-1β demonstrates cross talk between these pathways providing potential avenues for further research. This study highlights the potential negative effects of inflammation on muscle protein deposition and growth in fish and extends our understanding of energy allocation in ectothermic animals.
Collapse
Affiliation(s)
- Nicholas J Pooley
- Institute of Biological and Environmental Sciences, University of Aberdeen, Tillydrone Avenue, Aberdeen AB24 2TZ, UK
| | - Luca Tacchi
- Institute of Biological and Environmental Sciences, University of Aberdeen, Tillydrone Avenue, Aberdeen AB24 2TZ, UK
- Current address: Centre for Evolutionary and Theoretical Immunology, University of New Mexico, Albuquerque, NM 87131-0001, USA
| | - Christopher J Secombes
- Institute of Biological and Environmental Sciences, University of Aberdeen, Tillydrone Avenue, Aberdeen AB24 2TZ, UK
| | - Samuel AM Martin
- Institute of Biological and Environmental Sciences, University of Aberdeen, Tillydrone Avenue, Aberdeen AB24 2TZ, UK
| |
Collapse
|
21
|
Salem T, Gomard T, Court F, Moquet-Torcy G, Brockly F, Forné T, Piechaczyk M. Chromatin loop organization of the junb locus in mouse dendritic cells. Nucleic Acids Res 2013; 41:8908-25. [PMID: 23921639 PMCID: PMC3799436 DOI: 10.1093/nar/gkt669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The junb gene behaves as an immediate early gene in bacterial lipopolysaccharide (LPS)-stimulated dendritic cells (DCs), where its transient transcriptional activation is necessary for the induction of inflammatory cytokines. junb is a short gene and its transcriptional activation by LPS depends on the binding of NF-κB to an enhancer located just downstream of its 3′ UTR. Here, we have addressed the mechanisms underlying the transcriptional hyper-reactivity of junb. Using transfection and pharmacological assays to complement chromatin immunoprecipitation analyses addressing the localization of histones, polymerase II, negative elongation factor (NELF)-, DRB sensitivity-inducing factor (DSIF)- and Positive Transcription Factor b complexes, we demonstrate that junb is a RNA Pol II-paused gene where Pol II is loaded in the transcription start site domain but poorly active. Moreover, High salt-Recovered Sequence, chromosome conformation capture (3C)- and gene transfer experiments show that (i) junb is organized in a nuclear chromatin loop bringing into close spatial proximity the upstream promoter region and the downstream enhancer and (ii) this configuration permits immediate Pol II release on the junb body on binding of LPS-activated NF-κB to the enhancer. Thus, our work unveils a novel topological framework underlying fast junb transcriptional response in DCs. Moreover, it also points to a novel layer of complexity in the modes of action of NF-κB.
Collapse
Affiliation(s)
- Tamara Salem
- Equipe labellisée par la Ligue Nationale contre le Cancer, Institut de Génétique Moléculaire de Montpellier UMR 5535 CNRS, 1919 route de Mende, 34293 Montpellier cedex 5, France, Université Montpellier 2, Place Eugène Bataillon, 34095 Montpellier cedex 5, France and Université Montpellier 1, 5 Bd Henry IV, 34967 Montpellier cedex 2, France
| | | | | | | | | | | | | |
Collapse
|
22
|
Saben J, Zhong Y, Gomez-Acevedo H, Thakali KM, Borengasser SJ, Andres A, Shankar K. Early growth response protein-1 mediates lipotoxicity-associated placental inflammation: role in maternal obesity. Am J Physiol Endocrinol Metab 2013; 305:E1-14. [PMID: 23632636 PMCID: PMC4116409 DOI: 10.1152/ajpendo.00076.2013] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Obesity is associated with low-grade chronic inflammation, which contributes to cellular dysfunction promoting metabolic disease. Obesity during pregnancy leads to a proinflammatory milieu in the placenta; however, the underlying causes for obesity-induced placental inflammation remain unclear. Here, we examine the mechanisms by which saturated fatty acids and inflammatory cytokines induce inflammation in placental trophoblasts. We conducted global transcriptomic profiling in BeWo cells following palmitate and/or TNFα treatment and gene/protein expression analyses of MAPK pathways and characterized downstream transcription factors directly regulating inflammatory cytokines. Microarray analysis revealed increased expression of genes regulating inflammation, stress response, and immediate early response in cytotrophoblasts in response to palmitic acid (PA), TNFα, or a combination of both (PA + TNFα). Both gene ontology and gene set enrichment analysis revealed MAPK and EGR-1 signaling to be upregulated in BeWo cells, which was confirmed via immunoblotting. Importantly, activation of JNK signaling was necessary for increased proinflammatory cytokine (IL-6, TNFα, and IL-8) and EGR1 mRNA. Consistent with the requirement of JNK signaling, ChIP analysis confirmed the recruitment of c-Jun and other MAPK-responsive immediate early factors on the EGR1 promoter. Moreover, recruitment of EGR-1 on cytokine promoters (IL-6, TNFα, and IL-8) and an impaired proinflammatory response following knockdown of EGR-1 suggested it as a central component of the mechanism facilitating inflammatory gene expression. Finally, akin to in vitro findings, term placenta from obese women also had both increased JNK and p38 signaling and greater EGR-1 protein relative to lean women. Our results demonstrate that lipotoxic insults induce inflammation in placental cells via activation of JNK/EGR-1 signaling.
Collapse
Affiliation(s)
- Jessica Saben
- Arkansas Children's Nutrition Center, Little Rock, AR 72202, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Zhang G, Zhang Z, Liu Z. Interferon regulation factor-3 is a critical regulator of the mature of dendritic cells from mice. Scand J Immunol 2013; 77:13-20. [PMID: 23033912 DOI: 10.1111/sji.12005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 09/24/2012] [Indexed: 12/19/2022]
Abstract
Interferon regulatory factor-3 (IRF-3) plays an important role in virus and double-stranded RNA-mediated induction of type I interferon and RANTES (regulated on activation normal T cell expressed and secreted), DNA damage signalling, tumour suppression and virus-induced apoptosis. IRF-3 had recently been shown to contribute to T-cell activation in response to pathogens, which implicated an extensive immunological role for IRF-3. Dendritic cells (DCs) played critical roles as professional APCs in the development of immune responses. However, it was unclear whether IRF-3 had any effect on phenotype or function of DCs. In this study, it was shown that IRF-3 acted as a promoter of DC maturation. The level of IRF-3 expression was transiently upregulated and accumulated in the nucleus in TNF-α-induced immune maturation of mice DC cells. Knockdown of IRF-3 by small interfering RNA in DC cells resulted in both phenotypic and functional immaturation, even without TNF-α treatment. Overall, our data demonstrated for the first time that IRF-3 was a critical regulator of mice DC maturation.
Collapse
Affiliation(s)
- G Zhang
- Department of Hematology, Shengjing Hospital of China Medical University, Shenyang, China
| | | | | |
Collapse
|
24
|
Aberrant IKKα and IKKβ cooperatively activate NF-κB and induce EGFR/AP1 signaling to promote survival and migration of head and neck cancer. Oncogene 2013; 33:1135-47. [PMID: 23455325 PMCID: PMC3926900 DOI: 10.1038/onc.2013.49] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 01/07/2013] [Accepted: 01/18/2013] [Indexed: 12/23/2022]
Abstract
The Inhibitor-κB Kinase-Nuclear Factor-κB (IKK-NF-κB) and Epidermal Growth Factor Receptor-Activator Protein-1 (EGFR-AP-1) pathways are often co-activated and promote malignant behavior, but the underlying basis for this relationship is unclear. Resistance to inhibitors of IKKβ or EGFR is observed in head and neck squamous cell carcinomas (HNSCC). Here, we reveal that both IKKα and β contribute to nuclear activation of canonical and alternate NF-κB/REL family transcription factors, and overexpression of signal components enhancing co-activation of the EGFR-AP1 pathway. We observed that IKKα and IKKβ exhibit increased protein expression, nuclear localization and phosphorylation in HNSCC tissues and cell lines. Individually, IKK activity varied amongst different cell lines, but overexpression of both IKKs induced the strongest NF-κB activation. Conversely, siRNA knockdown of both IKKs significantly decreased nuclear localization and phosphorylation of canonical RELA and IκBα, and alternative p52 and RELB subunits. Knockdown of both IKKs more effectively inhibited NF-κB activation, broadly modulated gene expression, and suppressed cell proliferation and migration. Global expression profiling revealed that NF-κB, cytokine, inflammatory response, and growth factor signaling are among the top pathways and networks regulated by IKKs. Importantly, IKKα and IKKβ together promoted the expression and activity of TGFα, EGFR, and AP1 transcription factors cJun, JunB, and Fra1. Knockdown of AP1 subunits individually decreased 8/15 (53%) of IKK-targeted genes sampled, and similarly inhibited cell proliferation and migration. Mutations of NF-κB and AP1 binding sites abolished or decreased IKK-induced IL-8 promoter activity. Compounds such as wedelactone with dual IKK inhibitory activity, and geldanomycins that block IKKα/β and EGFR pathways were more active than IKKβ-specific inhibitors in suppressing NF-κB activation and proliferation, and inducing cell death. We conclude that IKKα and IKKβ cooperatively activate NF-κB and EGFR/AP1 networks of signaling pathways, and contribute to the malignant phenotype and the intrinsic or acquired therapeutic resistance of HNSCC.
Collapse
|
25
|
Tempé D, Vives E, Brockly F, Brooks H, De Rossi S, Piechaczyk M, Bossis G. SUMOylation of the inducible (c-Fos:c-Jun)/AP-1 transcription complex occurs on target promoters to limit transcriptional activation. Oncogene 2013; 33:921-7. [DOI: 10.1038/onc.2013.4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 12/15/2022]
|
26
|
Gao C, Liu Y, Ma L, Zhang X, Wang S. Effects of Ligustrazine on pulmonary damage in rats following scald injury. Burns 2012; 38:743-50. [DOI: 10.1016/j.burns.2011.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 11/25/2022]
|
27
|
Gao C, Liu Y, Ma L, Wang S. Protective effects of ulinastatin on pulmonary damage in rats following scald injury. Burns 2012; 38:1027-34. [PMID: 22455798 DOI: 10.1016/j.burns.2012.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 01/14/2012] [Accepted: 02/04/2012] [Indexed: 01/28/2023]
Abstract
Organ protection is desirable in severe burn/scald injuries, and damage mechanisms and thus effective therapies following scald injury have not been fully elucidated. Our aim was to examine the beneficial effects of ulinastatin on pulmonary damage associated with scald injury. Lewis rats were subjected to 30% total body surface area (TBSA) scald injury and were randomly divided into a burn control (S group) and an ulinastatin-treated group (U group). Lung malondialdehyde (MDA) and superoxide dismutase (SOD) levels were determined, and the lungs were examined histologically with immunohistochemistry (IHC) as well for the major histocompatibility complex (MHC) class I chain-related antigen A (MICA) and Bcl-2 at 24, 48 and 72 h after the injury. The expression of spleen human leucocyte antigen-DR (HLA-DR) was detected by immunohistochemistry analysis. Selectins and adhesion molecules in lungs and serum were also detected. The lung injury degree was represented as wet/dry (W/D) values and alveolar thickness. Ulinastatin decreased MDA levels and ameliorated the down-regulation of SOD activity. MICA was up-regulated after the scald, and this up-regulation was greatly diminished by ulinastatin. Bcl-2 was up-regulated after the scald, especially in the U group. The spleen HLA-DR expression demonstrated the immunoregulatory effects of ulinastatin, which effectively protected the pulmonary tissues from scald-induced injury. Our results demonstrated that pulmonary damage was associated with autoimmunity and oxidant attack after severe scald. Ulinastatin exhibits significant protective effects on these effects.
Collapse
Affiliation(s)
- Chengjin Gao
- Emergency Department and Surgical Intensive Care Unit, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | | | | | | |
Collapse
|
28
|
Shimp SK, Parson CD, Regna NL, Thomas AN, Chafin CB, Reilly CM, Nichole Rylander M. HSP90 inhibition by 17-DMAG reduces inflammation in J774 macrophages through suppression of Akt and nuclear factor-κB pathways. Inflamm Res 2012; 61:521-33. [PMID: 22327510 DOI: 10.1007/s00011-012-0442-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 12/14/2011] [Accepted: 01/23/2012] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE This study was designed to determine whether inhibition of heat shock protein 90 (HSP90) reduces pro-inflammatory mediator production by decreasing the nuclear factor (NF)-κB and Akt signaling pathways in immune-stimulated macrophages. METHODS J774A.1 murine macrophages were treated with the HSP90 inhibitor 17-DMAG (0.01, 0.1 or 1 μM) prior to immune stimulation with lipopolysaccharide and interferon-γ. Expression of Akt, inhibitor of κB kinase (IKK), and heat shock proteins were measured in whole cell lysates by Western blotting. Phosphorylated Akt and inhibitor of κB (IκB) were measured in whole cell lysates by ELISA. Cell supernatants were analyzed for interleukin (IL)-6, tumor necrosis factor (TNF)-α and nitric oxide (NO). Translocation of NF-κB and heat shock factor (HSF)-1 was assessed by immunofluorescence. RESULTS Treating cells with 17-DMAG reduced expression of Akt and IKK in immune-stimulated cells. 17-DMAG reduced nuclear translocation of NF-κB and reduced immune-stimulated production of IL-6, TNF-α and NO, but did not decrease inducible nitric oxide synthase expression. CONCLUSIONS Our studies show that the immune-mediated NF-κB inflammatory cascade is blocked by the HSP90 inhibitor 17-DMAG. Due to the broad interaction of HSP90 with many pro-inflammatory kinase cascades, inhibition of HSP90 may provide a novel approach to reducing chronic inflammation.
Collapse
Affiliation(s)
- Samuel K Shimp
- Virginia Polytechnic Institute and State University, Virginia Tech-Wake Forest School of Biomedical Engineering and Science, Blacksburg, VA 24061, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Carignan D, Désy O, de Campos-Lima PO. The dysregulation of the monocyte/macrophage effector function induced by isopropanol is mediated by the defective activation of distinct members of the AP-1 family of transcription factors. Toxicol Sci 2011; 125:144-56. [PMID: 22020770 DOI: 10.1093/toxsci/kfr283] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Isopropanol is the second most common cause of short-chain alcohol acute intoxication. Nonethanolic short-chain alcohols mediate their immunomodulatory effect by interfering with nuclear factor of activated T cells (NFAT) activation with or without additional activator protein-1 (AP-1) involvement. In the present study, we examined the immunomodulation induced by isopropanol in conditions that are not reliant on NFAT: the inflammatory cytokine response of lipopolysaccharide (LPS)-stimulated monocytes. Our hypothesis was that isopropanol acute exposure would have an attenuated effect or no consequence in this setting. To our surprise, the impairment of AP-1 activation was sufficient to mediate a severe and dose-dependent phenotype in human monocytes in vitro at alcohol concentrations as low as 0.16% (or 26 mM). There were three outcomes: interleukin (IL)-1β/IL-8 were unaltered; IL-6 was upregulated; and tumor necrosis factor alpha (TNF-α)/CCL2 were downregulated. The effector function of human monocyte-derived macrophages was also compromised. Our results showed that Toll-like receptor 4 early signaling was preserved, as isopropanol did not change the kinase activity of the IL-1 receptor-associated kinase 1 in LPS-stimulated cells. The nuclear factor-κB signaling cascade and the p38/c-Jun N-terminal kinase modules of the mitogen-activated protein kinase pathway were alcohol insensitive. Conversely, the activation of extracellular signal-regulated protein kinase and, ultimately, of c-Fos and JunB were impaired. The alcohol-induced cytokine dysregulation was confirmed in a mouse model of isopropanol intoxication in which the production of TNF-α in response to LPS challenge was virtually abolished. The magnitude of this alcohol effect was sufficiently high to rescue animals from LPS-induced toxic shock. Our data contribute to the dismal body of information on the immunotoxicology of isopropanol, one of the most ubiquitous chemicals to which the general population is significantly exposed.
Collapse
Affiliation(s)
- Damien Carignan
- Laval University Cancer Research Center, Quebec City, Quebec G1R 2J6, Canada
| | | | | |
Collapse
|
30
|
|
31
|
Ghannam S, Pène J, Torcy-Moquet G, Jorgensen C, Yssel H. Mesenchymal Stem Cells Inhibit Human Th17 Cell Differentiation and Function and Induce a T Regulatory Cell Phenotype. THE JOURNAL OF IMMUNOLOGY 2010; 185:302-12. [DOI: 10.4049/jimmunol.0902007] [Citation(s) in RCA: 412] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|