1
|
Mishra S, Welch N, Singh SS, Singh KD, Bellar A, Kumar A, Deutz LN, Hanlon MD, Kant S, Dastidar S, Patel H, Agrawal V, Attaway AH, Musich R, Stark GR, Tedesco FS, Truskey GA, Weiner ID, Karnik SS, Dasarathy S. Ammonia transporter RhBG initiates downstream signaling and functional responses by activating NFκB. Proc Natl Acad Sci U S A 2024; 121:e2314760121. [PMID: 39052834 PMCID: PMC11294993 DOI: 10.1073/pnas.2314760121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 06/19/2024] [Indexed: 07/27/2024] Open
Abstract
Transceptors, solute transporters that facilitate intracellular entry of molecules and also initiate intracellular signaling events, have been primarily studied in lower-order species. Ammonia, a cytotoxic endogenous metabolite, is converted to urea in hepatocytes for urinary excretion in mammals. During hyperammonemia, when hepatic metabolism is impaired, nonureagenic ammonia disposal occurs primarily in skeletal muscle. Increased ammonia uptake in skeletal muscle is mediated by a membrane-bound, 12 transmembrane domain solute transporter, Rhesus blood group-associated B glycoprotein (RhBG). We show that in addition to its transport function, RhBG interacts with myeloid differentiation primary response-88 (MyD88) to initiate an intracellular signaling cascade that culminates in activation of NFκB. We also show that ammonia-induced MyD88 signaling is independent of the canonical toll-like receptor-initiated mechanism of MyD88-dependent NFκB activation. In silico, in vitro, and in situ experiments show that the conserved cytosolic J-domain of the RhBG protein interacts with the Toll-interleukin-1 receptor (TIR) domain of MyD88. In skeletal muscle from human patients, human-induced pluripotent stem cell-derived myotubes, and myobundles show an interaction of RhBG-MyD88 during hyperammonemia. Using complementary experimental and multiomics analyses in murine myotubes and mice with muscle-specific RhBG or MyD88 deletion, we show that the RhBG-MyD88 interaction is essential for the activation of NFkB but not ammonia transport. Our studies show a paradigm of substrate-dependent regulation of transceptor function with the potential for modulation of cellular responses in mammalian systems by decoupling transport and signaling functions of transceptors.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Nicole Welch
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
- Gastroenterology and Hepatology, Lerner Research Institute, Cleveland, OH44195
| | - Shashi Shekhar Singh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | | | - Annette Bellar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Avinash Kumar
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Lars N. Deutz
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Maxmillian D. Hanlon
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Sashi Kant
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Sumitava Dastidar
- Department of Cell and Developmental Biology, University College London & The Francis Crick Institute, LondonWC1E6DE, UK
| | - Hailee Patel
- Duke Biomedical Engineering, Duke University, Durham, NC27708
| | - Vandana Agrawal
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - Amy H. Attaway
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
- Pulmonary Medicine, Lerner Research Institute, Cleveland, OH44195
| | - Ryan Musich
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
| | - George R. Stark
- Cancer Biology, Lerner Research Institute, Cleveland, OH44195
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London & The Francis Crick Institute, LondonWC1E6DE, UK
| | | | - I. David Weiner
- Division of Nephrology Hypertension, and Renal Transplantation, University of Florida, Gainesville, FL32610
- Nephrology and Hypertension Section, Gainesville, FL32610
| | - Sadashiva S. Karnik
- Cardiovascular and Metabolic Diseases, Lerner Research Institute, Cleveland, OH44195
| | - Srinivasan Dasarathy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland, OH44195
- Gastroenterology and Hepatology, Lerner Research Institute, Cleveland, OH44195
| |
Collapse
|
2
|
Putnam CM, Kondeti L, Kesler MBA, Varney ME. Modulating the immune system as a therapeutic target for myelodysplastic syndromes and acute myeloid leukemia. Biochem Cell Biol 2023; 101:481-495. [PMID: 37566901 DOI: 10.1139/bcb-2022-0374] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2023] Open
Abstract
Modulating the immune system to treat diseases, including myeloid malignancies, has resulted in the development of a multitude of novel therapeutics in recent years. Myelodysplastic syndromes or neoplasms (MDS) and acute myeloid leukemia (AML) are hematologic malignancies that arise from defects in hematopoietic stem and progenitor cells (HSPCs). Dysregulated immune responses, especially in innate immune and inflammatory pathways, are highly associated with the acquisition of HSPC defects in MDS and AML pathogenesis. In addition to utilizing the immune system in immunotherapeutic interventions such as chimeric antigen receptor T cell therapy, vaccines, and immune checkpoint inhibitors, mitigating dysregulation of innate immune and inflammatory responses in MDS and AML remains a priority in slowing the initiation and progression of these myeloid malignancies. This review provides a comprehensive summary of the current progress of diverse strategies to utilize or modulate the immune system in the treatment of MDS and AML.
Collapse
Affiliation(s)
- Caroline M Putnam
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Lahari Kondeti
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Meredith B A Kesler
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| | - Melinda E Varney
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV, USA
| |
Collapse
|
3
|
Wang L, Cui Y, Liu H, Wu J, Li J, Liu X. PM2.5 aggravates airway inflammation in asthmatic mice: activating NF-κB via MyD88 signaling pathway. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2023; 33:563-574. [PMID: 35227140 DOI: 10.1080/09603123.2022.2041561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The role of PM2.5 in the bronchial asthma remains unclear. In this study, the deficient mice of TLR4-/-, TLR2-/- and MyD88 -/- were used to establish asthma model. The effects of PM2.5 on the inflammatory response in lung tissue of these mice were observed. PM2.5 increased alveolar macrophages and neutrophils, up-regulated the IL-12 and KC expression in WT mice, but down-regulated their levels in TLR2 -/-, TLR4 -/- and MyD88 -/- mice. OVA+PM2.5 stimulated neutrophil count in WT mice, but it decreased in TLR2 -/- and TLR4 -/- mice. OVA+PM2.5 also increased the Eotaxin, IL-5, IL-13 and MCP-3 expression levels, and OVA specific IgE and IgG1 in serum also increased in WT group. PM2.5 may activate NF-κB through the TLR2/TLR4/MyD88 signaling pathway and aggravate allergic inflammation of lung in asthmatic mice. The microelements in PM2.5 granules, such as lipopolysaccharide, may be an important factor in the high incidence of asthma.
Collapse
Affiliation(s)
- Lei Wang
- Department of Respiratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Yanzhi Cui
- Department of Respiratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Hu Liu
- Department of Respiratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Jing Wu
- Department of Respiratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Jie Li
- Department of Respiratory Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi, China
| | - Xiansheng Liu
- Department of Respiratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
4
|
Reus JB, Rex EA, Gammon DB. How to Inhibit Nuclear Factor-Kappa B Signaling: Lessons from Poxviruses. Pathogens 2022; 11:pathogens11091061. [PMID: 36145493 PMCID: PMC9502310 DOI: 10.3390/pathogens11091061] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
The Nuclear Factor-kappa B (NF-κB) family of transcription factors regulates key host inflammatory and antiviral gene expression programs, and thus, is often activated during viral infection through the action of pattern-recognition receptors and cytokine–receptor interactions. In turn, many viral pathogens encode strategies to manipulate and/or inhibit NF-κB signaling. This is particularly exemplified by vaccinia virus (VV), the prototypic poxvirus, which encodes at least 18 different inhibitors of NF-κB signaling. While many of these poxviral NF-κB inhibitors are not required for VV replication in cell culture, they virtually all modulate VV virulence in animal models, underscoring the important influence of poxvirus–NF-κB pathway interactions on viral pathogenesis. Here, we review the diversity of mechanisms through which VV-encoded antagonists inhibit initial NF-κB pathway activation and NF-κB signaling intermediates, as well as the activation and function of NF-κB transcription factor complexes.
Collapse
|
5
|
Canaria DA, Clare MG, Yan B, Campbell CB, Ismaio ZA, Anderson NL, Park S, Dent AL, Kazemian M, Olson MR. IL-1β promotes IL-9-producing Th cell differentiation in IL-2-limiting conditions through the inhibition of BCL6. Front Immunol 2022; 13:1032618. [PMID: 36389679 PMCID: PMC9663844 DOI: 10.3389/fimmu.2022.1032618] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022] Open
Abstract
IL-9-producing CD4+ T helper cells, termed Th9 cells, differentiate from naïve precursor cells in response to a combination of cytokine and cell surface receptor signals that are elevated in inflamed tissues. After differentiation, Th9 cells accumulate in these tissues where they exacerbate allergic and intestinal disease or enhance anti-parasite and anti-tumor immunity. Previous work indicates that the differentiation of Th9 cells requires the inflammatory cytokines IL-4 and TGF-β and is also dependent of the T cell growth factor IL-2. While the roles of IL-4 and TGF-β-mediated signaling are relatively well understood, how IL-2 signaling contributes to Th9 cell differentiation outside of directly inducing the Il9 locus remains less clear. We show here that murine Th9 cells that differentiate in IL-2-limiting conditions exhibit reduced IL-9 production, diminished NF-kB activation and a reduced NF-kB-associated transcriptional signature, suggesting that IL-2 signaling is required for optimal NF-kB activation in Th9 cells. Interestingly, both IL-9 production and the NF-kB transcriptional signature could be rescued by addition of the NF-kB-activating cytokine IL-1β to IL-2-limiting cultures. IL-1β was unique among NF-kB-activating factors in its ability to rescue Th9 differentiation as IL-2 deprived Th9 cells selectively induced IL-1R expression and IL-1β/IL-1R1 signaling enhanced the sensitivity of Th9 cells to limiting amounts of IL-2 by suppressing expression of the Th9 inhibitory factor BCL6. These data shed new light on the intertwined nature of IL-2 and NF-kB signaling pathways in differentiating Th cells and elucidate the potential mechanisms that promote Th9 inflammatory function in IL-2-limiting conditions.
Collapse
Affiliation(s)
- D Alejandro Canaria
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Maia G Clare
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Bingyu Yan
- Department of Biochemistry, Purdue University, West Lafayette, IN, United States
| | - Charlotte B Campbell
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Zachariah A Ismaio
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Nicole L Anderson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Sungtae Park
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| | - Alexander L Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Majid Kazemian
- Department of Biochemistry, Purdue University, West Lafayette, IN, United States.,Department of Computer Science, Purdue University, West Lafayette, IN, United States
| | - Matthew R Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
6
|
Rashidi S, Mansouri R, Ali-Hassanzadeh M, Ghani E, Barazesh A, Karimazar M, Nguewa P, Carrera Silva EA. Highlighting the interplay of microRNAs from Leishmania parasites and infected-host cells. Parasitology 2021; 148:1434-1446. [PMID: 34218829 PMCID: PMC11010138 DOI: 10.1017/s0031182021001177] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 06/10/2021] [Accepted: 06/27/2021] [Indexed: 02/05/2023]
Abstract
Leishmania parasites, the causative agents of leishmaniasis, are protozoan parasites with the ability to modify the signalling pathway and cell responses of their infected host cells. These parasite strategies alter the host cell environment and conditions favouring their replication, survival and pathogenesis. Since microRNAs (miRNAs) are able to post-transcriptionally regulate gene expression processes, these biomolecules can exert critical roles in controlling Leishmania-host cell interplay. Therefore, the identification of relevant miRNAs differentially expressed in Leishmania parasites as well as in infected cells, which affect the host fitness, could be critical to understand the infection biology, pathogenicity and immune response against these parasites. Accordingly, the current review aims to address the differentially expressed miRNAs in both, the parasite and infected host cells and how these biomolecules change cell signalling and host immune responses during infection. A deep understanding of these processes could provide novel guidelines and therapeutic strategies for managing and treating leishmaniasis.
Collapse
Affiliation(s)
- Sajad Rashidi
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Mansouri
- Department of Immunology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences and Health Services, Yazd, Iran
| | - Mohammad Ali-Hassanzadeh
- Department of Immunology, School of Medicine, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Esmaeel Ghani
- Endocrinology and Metabolism Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Afshin Barazesh
- Department of Microbiology and Parasitology, Faculty of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mohammadreza Karimazar
- Department of Parasitology and Mycology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Paul Nguewa
- University of Navarra, ISTUN Instituto de Salud Tropical, Department of Microbiology and Parasitology, IdiSNA (Navarra Institute for Health Research), c/Irunlarrea 1, 31008Pamplona, Spain
| | | |
Collapse
|
7
|
Sanders OD, Rajagopal L, Rajagopal JA. Does oxidatively damaged DNA drive amyloid-β generation in Alzheimer's disease? A hypothesis. J Neurogenet 2021; 35:351-357. [PMID: 34282704 DOI: 10.1080/01677063.2021.1954641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
In Alzheimer's disease (AD), amyloid-β (Aβ) generation and upstream β-secretase 1 (BACE1) expression appear to be driven by oxidative stress via c-Jun N-terminal kinase (JNK), p38, and Interferon-Induced, Double-Stranded RNA-Activated Protein Kinase (PKR). In addition, inflammatory molecules, including lipopolysaccharide (LPS), induce genes central to Aβ genesis, such as BACE1, via nuclear factor-κB (NFκB). However, additional triggers of Aβ generation remain poorly understood and might represent novel opportunities for therapeutic intervention. Based on mechanistic studies and elevated ectopic oxidatively damaged DNA (oxoDNA) levels in preclinical AD, mild cognitive impairment, and AD patients, we hypothesize oxoDNA contributes to β-amyloidosis starting from the earliest stages of AD through multiple pathways. OxoDNA induces mitogen-activated protein kinase kinase kinase kinase 4 (MAP4K4), thereby sensitizing the brain to oxidative stress-induced JNK activation and BACE1 transcription. It also induces myeloid differentiation primary response 88 (MyD88) and activates protein kinase CK2, thereby increasing NFκB activation and BACE1 induction. OxoDNA increases oxidative stress via nuclear factor erythroid 2-related factor 2 (Nrf2) ectopic localization, likely augmenting JNK-mediated BACE1 induction. OxoDNA likely also promotes β-amyloidosis via absent in melanoma 2 (AIM2) induction. Falsifiable predictions of this hypothesis include that deoxyribonuclease treatment should decrease Aβ and possibly slow cognitive decline in AD patients. While formal testing of this hypothesis remains to be performed, a case report has found deoxyribonuclease I treatment improved a severely demented AD patient's Mini-Mental Status Exam score from 3 to 18 at 2 months. There is preliminary preclinical and clinical evidence suggesting that ectopic oxidatively damaged DNA may act as an inflammatory damage-associated molecular pattern contributing to Aβ generation in AD, and deoxyribonuclease I should be formally evaluated to test whether it can decrease Aβ levels and slow cognitive decline in AD patients.
Collapse
|
8
|
Das S, Mukherjee S, Ali N. Super enhancer-mediated transcription of miR146a-5p drives M2 polarization during Leishmania donovani infection. PLoS Pathog 2021; 17:e1009343. [PMID: 33630975 PMCID: PMC7943006 DOI: 10.1371/journal.ppat.1009343] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 03/09/2021] [Accepted: 01/28/2021] [Indexed: 12/13/2022] Open
Abstract
The outcome of Leishmania donovani infection depends upon the dynamic interchanges between M1 and M2 macrophages. Information of the involvement of microRNAs (miRNAs) and epigenetic modifiers in regulating macrophage plasticity during L. donovani infection is still elusive. Differential expression analysis of polarization-regulating miRNAs, revealed significant enrichment of miR146a-5p during Leishmania donovani infection. A sustained enrichment of miR146a-5p was observed in both infected bone marrow derived macrophages (BMDMs) and BALB/c mice organs. We found involvement of miR146a-5p in phagocytosis and survivability of parasites. Moreover, miR146a-5pgot enriched in interleukin 4- stimulated BMDMs, indicating its possible involvement in M2 polarization. Upon transfecting BMDMs with miRVANA anti-146a oligos, M2 markers (CCR7, YM-1, FIZZ-1, arginase-1, IL10 and IL4) and transcription factors (p-STAT6 and c/EBPβ) got depleted with concomitant augmentation of M1-polarizing transcription factors (p-STAT1, AP1 and IRF-1), miR146a target genes (TRAF6 and IRAK1), M1 cytokines (IL12 and TNFα), iNOS, nitric oxide, and nuclear translocation of phospho p-65 subunit. Neutralization of intracellular mature miR146a-5p pool in infected BALB/c mice lower organ parasite burden and expressions of M2 markers and IL10 with enrichment of M1 markers like iNOS and IL12. Additionally, we explored the novel role of super enhancer (SE), a cis-acting regulatory component, to enrich miR146a-5p expression during infection. Enhanced expression and nuclear retention of SE components like BET bromodomain 4 (BRD4) and p300 were found in infected BMDMs. Upon silencing BRD4, expressions of miR146a-5p and M2 markers were down regulated and TRAF6, IRAK1 and iNOS levels increased. STRING V.11 based predication and immune precipitation confirmed the strong interaction amongst BRD4, p300 and RNA pol II (RpbI). Chromatin immune precipitation studies suggested the recruitment of BRD4 at the enhancer loci of miR146a-5p gene during infection. Altogether, our findings revealed a novel role of BRD4/p300-depdendent super-enhancer in regulating miR146a expression during L. donovani infection which in turn mediates M2 polarization and immune-suppression. Visceral leishmaniasis (VL), caused by protozoan parasites Leishmania donovani, is the most severe form of leishmaniasis and is highly lethal if left untreated. Major obstacle for successful therapy of VL originates from the life-long immune-suppression triggered in the post kala-azar dermal leishmaniasis (PKDL) patients during infection. Identification of molecular principles behind such immune-suppression will add success in VL therapeutics. L. donovani hijacks the host macrophages and converts them from pro-inflammatory M1 to immune-suppressive M2 type, which allows successful infection establishment. Herein, we explored the indispensable role of miRNA-146a-5p in conversion of M1 to M2 type during infection. Both in vitro and in vivo miRNA silencing established miR146a-5p as an imperative negative regulator ofM1 polarization. Computational analysis as well as immune precipitation based experiments authenticated that L. donovani induces super enhancer complex mediated transcriptional upregulation of miR146a-5p. BET bromodomain protein 4 (BRD4) forms this SE complex along with p300 histone acetyl transferase and RNA pol II. Silencing of BRD4 significantly abrogated miR146a-5p mediated M2 polarization. In short, our current findings established a previously unrecognized role of BRD4-depdendent super enhancers in orchestrating persistent transcription of macrophage miR146a-5p which in turn promotes M2 polarization during L. donovani infection.
Collapse
Affiliation(s)
- Sonali Das
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sohitri Mukherjee
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Nahid Ali
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- * E-mail:
| |
Collapse
|
9
|
Zgórzyńska E, Stulczewski D, Dziedzic B, Su KP, Walczewska A. Docosahexaenoic fatty acid reduces the pro-inflammatory response induced by IL-1β in astrocytes through inhibition of NF-κB and AP-1 transcription factor activation. BMC Neurosci 2021; 22:4. [PMID: 33499800 PMCID: PMC7839194 DOI: 10.1186/s12868-021-00611-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 01/08/2021] [Accepted: 01/13/2021] [Indexed: 01/02/2023] Open
Abstract
Background Astrocytes are responsible for a broad range of functions that maintain homeostasis in the brain. However, their response to the pro-inflammatory cytokines released by activated microglia in various neurological pathologies may exacerbate neurodegenerative processes. Accumulating evidence suggests that omega-3 docosahexaenoic fatty acid (DHA) has an anti-inflammatory effect in various cell cultures studies and in a variety of neurological disorders. In this study we examined the mechanism involved in the inhibition of the pro-inflammatory response by DHA in astrocytes treated with IL-1β. Methods and results Activation of the transcription factors NF-κB and AP-1 was measured in IL-1β-treated primary astrocytes incubated with various concentrations of DHA. COX-2 and iNOS protein expression was determined by Western blot, and TNF-α and IL-6 secretion was measured using ELISA-based assays. DHA treatment inhibited translocation of p65NF-κB to the nucleus, significantly lowered p65NF-κB protein level and fluorescence of p65NF-κB in the nucleus, reduced dose-dependently IκB protein phosphorylation, and the binding of the AP-1 transcription factor members (c-Jun/c-Fos) to the specific TPA-response element (TRE) of DNA. In addition, the expression of pro-inflammatory COX-2 and iNOS proteins was downregulated and TNF-α and IL-6 secretion was also reduced. Conclusions These results indicate that DHA is a powerful factor that reduces the pro-inflammatory response in astrocytes. Consequently, successful introduction of DHA into the astrocyte membranes can attenuate neuroinflammation, which is a key factor of age-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Emilia Zgórzyńska
- Department of Cell-to-Cell Communication, Medical University of Lodz, Mazowiecka 6/8, 92- 215, Lodz, Poland.
| | - Dawid Stulczewski
- Department of Cell-to-Cell Communication, Medical University of Lodz, Mazowiecka 6/8, 92- 215, Lodz, Poland
| | - Barbara Dziedzic
- Department of Cell-to-Cell Communication, Medical University of Lodz, Mazowiecka 6/8, 92- 215, Lodz, Poland
| | - Kuan-Pin Su
- An-Nan Hospital, China Medical University, Tainan, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan.,Department of Psychiatry and Mind-Body Interface Laboratory, China Medical University, Taichung, Taiwan
| | - Anna Walczewska
- Department of Cell-to-Cell Communication, Medical University of Lodz, Mazowiecka 6/8, 92- 215, Lodz, Poland
| |
Collapse
|
10
|
The NLRP3 inflammasome regulates adipose tissue metabolism. Biochem J 2020; 477:1089-1107. [PMID: 32202638 DOI: 10.1042/bcj20190472] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/27/2022]
Abstract
Adipose tissue regulates metabolic homeostasis by participating in endocrine and immune responses in addition to storing and releasing lipids from adipocytes. Obesity skews adipose tissue adipokine responses and degrades the coordination of adipocyte lipogenesis and lipolysis. These defects in adipose tissue metabolism can promote ectopic lipid deposition and inflammation in insulin-sensitive tissues such as skeletal muscle and liver. Sustained caloric excess can expand white adipose tissue to a point of maladaptation exacerbating both local and systemic inflammation. Multiple sources, instigators and propagators of adipose tissue inflammation occur during obesity. Cross-talk between professional immune cells (i.e. macrophages) and metabolic cells (i.e. adipocytes) promote adipose tissue inflammation during metabolic stress (i.e. metaflammation). Metabolic stress and endogenous danger signals can engage pathogen recognition receptors (PRRs) of the innate immune system thereby activating pro-inflammatory and stress pathways in adipose tissue. The Nod-like receptor protein 3 (NLRP3) inflammasome can act as a metabolic danger sensor to a wide range of pathogen- and damage-associated molecular patterns (PAMPs and DAMPs). Activation of the NLRP3 inflammasome facilitates caspase-1 dependent production of the pro-inflammatory cytokines IL-1β and IL-18. Activation of the NLRP3 inflammasome can promote inflammation and pyroptotic cell death, but caspase-1 is also involved in adipogenesis. This review discusses the role of the NLRP3 inflammasome in adipose tissue immunometabolism responses relevant to metabolic disease. Understanding the potential sources of NLRP3 activation and consequences of NLRP3 effectors may reveal therapeutic opportunities to break or fine-tune the connection between metabolism and inflammation in adipose tissue during obesity.
Collapse
|
11
|
Human Metapneumovirus Small Hydrophobic Protein Inhibits Interferon Induction in Plasmacytoid Dendritic Cells. Viruses 2018; 10:v10060278. [PMID: 29789500 PMCID: PMC6024365 DOI: 10.3390/v10060278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 05/20/2018] [Accepted: 05/21/2018] [Indexed: 12/21/2022] Open
Abstract
Human metapneumovirus (hMPV), a leading cause of respiratory tract infections in infants, encodes a small hydrophobic (SH) protein of unknown function. Here we show that infection of plasmacytoid dendritic cells (pDCs) with a recombinant virus lacking SH expression (rhMPV-ΔSH) enhanced the secretion of type I interferons (IFNs), which required TLR7 and MyD88 expression. HMPV SH protein inhibited TLR7/MyD88/TRAF6 signaling leading to IFN gene transcription, identifying a novel mechanism by which paramyxovirus SH proteins modulate innate immune responses.
Collapse
|
12
|
Tsuchiya M, Sekiai S, Hatakeyama H, Koide M, Chaweewannakorn C, Yaoita F, Tan-No K, Sasaki K, Watanabe M, Sugawara S, Endo Y, Itoi E, Hagiwara Y, Kanzaki M. Neutrophils Provide a Favorable IL-1-Mediated Immunometabolic Niche that Primes GLUT4 Translocation and Performance in Skeletal Muscles. Cell Rep 2018; 23:2354-2364. [DOI: 10.1016/j.celrep.2018.04.067] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 03/09/2018] [Accepted: 04/14/2018] [Indexed: 11/27/2022] Open
|
13
|
Li Y, Wang Y, Zhou L, Liu M, Liang G, Yan R, Jiang Y, Hao J, Zhang X, Hu X, Huang Y, Wang R, Yin Z, Wu J, Luo G, He W. Vγ4 T Cells Inhibit the Pro-healing Functions of Dendritic Epidermal T Cells to Delay Skin Wound Closure Through IL-17A. Front Immunol 2018; 9:240. [PMID: 29483920 PMCID: PMC5816340 DOI: 10.3389/fimmu.2018.00240] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Abstract
Dendritic epidermal T cells (DETCs) and dermal Vγ4 T cells engage in wound re-epithelialization and skin inflammation. However, it remains unknown whether a functional link between Vγ4 T cell pro-inflammation and DETC pro-healing exists to affect the outcome of skin wound closure. Here, we revealed that Vγ4 T cell-derived IL-17A inhibited IGF-1 production by DETCs to delay skin wound healing. Epidermal IL-1β and IL-23 were required for Vγ4 T cells to suppress IGF-1 production by DETCs after skin injury. Moreover, we clarified that IL-1β rather than IL-23 played a more important role in inhibiting IGF-1 production by DETCs in an NF-κB-dependent manner. Together, these findings suggested a mechanistic link between Vγ4 T cell-derived IL-17A, epidermal IL-1β/IL-23, DETC-derived IGF-1, and wound-healing responses in the skin.
Collapse
Affiliation(s)
- Yashu Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yangping Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lina Zhou
- Department of Endocrinology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Meixi Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| | - Guangping Liang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rongshuai Yan
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yufeng Jiang
- Wound Healing Center, Medical School of Chinese PLA, Chinese PLA General Hospital, Beijing, China
| | - Jianlei Hao
- Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| | - Yong Huang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| | - Rupeng Wang
- Department of Dermatology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Guangdong Province Key Laboratory of Molecular Immunology and Antibody Engineering, Jinan University, Guangzhou, China
| | - Jun Wu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China.,Department of Burns, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory for Disease Proteomics, Department of Endocrinology of Southwest Hospital, Chongqing, China
| |
Collapse
|
14
|
Xu Y, Huang Y, Cai S. Characterization and function analysis of interleukin-1 receptor-associated kinase-1 (IRAK-1) from Fenneropenaeus penicillatus. FISH & SHELLFISH IMMUNOLOGY 2017; 61:111-119. [PMID: 28025158 DOI: 10.1016/j.fsi.2016.12.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 12/15/2016] [Accepted: 12/22/2016] [Indexed: 06/06/2023]
Abstract
The interleukin-1 receptor-associated kinase-1 (IRAK-1) is an important adapter protein which links downstream of MyD88, and involved in the complex composed of MyD88 and TRAF6 to activate TLRs signaling pathway. In this study, an IRAK-1 homolog (FpIRAK-1) was cloned from the red tail shrimp Fenneropenaeus penicillatus. The ORF of FpIRAK-1 consisted of 2874 bp encoding a protein of 957 amino acids which contains a death domain (DD) and a catalytic domain of serine/threonine kinases (STKc). Homology analysis revealed that the predicted amino acid sequence of FpIRAK-1 shared 71% similarities with IRAK-1 of Litopenaeus vannamei. Real-time RT-PCR indicated that FpIRAK-1 was constitutively expressed in various tissues of F. penicillatus. The expression level of FpIRAK-1 mRNA was significantly up-regulated and then decreased gradually after white spot syndrome virus (WSSV) and Vibrio alginolyticus challenge. Gene knockdown of FpIRAK-1 enhanced the sensitivity of shrimps to WSSV and V. alginolyticus challenge, suggesting FpIRAK-1 could play a positive role against bacterial and viral pathogens. In conclusion, the results of this study provide some insights into the function of FpIRAK-1 in activating Toll signaling pathway and the host defense against invading pathogens.
Collapse
Affiliation(s)
- Youhou Xu
- Guangxi Key Laboratory of Beibu Gulf Marine Biodiversity Conservation, Qinzhou University, Qinzhou, China
| | - Yucong Huang
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institutes, Fisheries College of Guangdong Ocean University, Zhanjiang, China
| | - Shuanghu Cai
- Guangdong Provincial Key Laboratory of Pathogenic Biology and Epidemiology for Aquatic Economic Animals & Key Laboratory of Control for Diseases of Aquatic Economic Animals of Guangdong Higher Education Institutes, Fisheries College of Guangdong Ocean University, Zhanjiang, China.
| |
Collapse
|
15
|
The modern interleukin-1 superfamily: Divergent roles in obesity. Semin Immunol 2016; 28:441-449. [DOI: 10.1016/j.smim.2016.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/03/2016] [Accepted: 10/03/2016] [Indexed: 11/20/2022]
|
16
|
Algate K, Haynes DR, Bartold PM, Crotti TN, Cantley MD. The effects of tumour necrosis factor-α on bone cells involved in periodontal alveolar bone loss; osteoclasts, osteoblasts and osteocytes. J Periodontal Res 2015; 51:549-66. [DOI: 10.1111/jre.12339] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2015] [Indexed: 12/22/2022]
Affiliation(s)
- K. Algate
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - D. R. Haynes
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - P. M. Bartold
- School of Dentistry; University of Adelaide; Adelaide SA Australia
| | - T. N. Crotti
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
| | - M. D. Cantley
- Discipline of Anatomy and Pathology; University of Adelaide; Adelaide SA Australia
- Myeloma Research Laboratory; University of Adelaide; Adelaide SA Australia
| |
Collapse
|
17
|
Islam SU, Shehzad A, Lee YS. Prostaglandin E2inhibits resveratrol-induced apoptosis through activation of survival signaling pathways in HCT-15 cell lines. Anim Cells Syst (Seoul) 2015. [DOI: 10.1080/19768354.2015.1101398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
18
|
Mukherjee B, Paul J, Mukherjee S, Mukhopadhyay R, Das S, Naskar K, Sundar S, Dujardin JC, Saha B, Roy S. Antimony-Resistant Leishmania donovani Exploits miR-466i To Deactivate Host MyD88 for Regulating IL-10/IL-12 Levels during Early Hours of Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:2731-42. [PMID: 26283478 DOI: 10.4049/jimmunol.1402585] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 07/15/2015] [Indexed: 01/29/2023]
Abstract
Infection with antimony-resistant Leishmania donovani (Sb(R)LD) induces aggressive pathology in the mammalian hosts as compared with ones with antimony-sensitive L. donovani (Sb(S)LD) infection. Sb(R)LD, but not Sb(S)LD, interacts with TLR2/TLR6 to induce IL-10 by exploiting p50/c-Rel subunits of NF-κB in infected macrophages (Mϕs). Most of the TLRs exploit the universal adaptor protein MyD88 to activate NF-κB. We now show that infection of Mϕs from MyD88(-/-) mice with Sb(R)LD gave rise to significantly higher intracellular parasite number coupled with elevated IL-10/IL-12 ratio in the culture supernatant as compared with infection in wild type (WT) Mϕs. Τhese attributes were not seen with Sb(S)LD in similar experiments. Further, Sb(R)LD infection upregulated miR-466i, which binds with 3'-untranslated region, leading to the downregulation of MyD88. Infection of MyD88(-/-) Mϕ or IL-12(-/-) Mϕ with Sb(R)LD induced IL-10 surge at 4 h, whereas the same in WT Mϕ started from 12 h. Thus, absence of IL-12 in MyD88(-/-) mice favored early binding of NF-κB subunits to the IL-10 promoter, resulting in IL-10 surge. Infection of MyD88(-/-) mice with Sb(R)LD showed significantly higher organ parasites coupled with ill-defined and immature hepatic granulomas, whereas in WT mice there were less organ parasites and the granulomas were well defined. From the survival kinetics it was observed that Sb(R)LD-infected MyD88(-/-) mice died by 60 d postinfection, whereas the WT mice continued to survive. Our results demonstrate that Sb(R)LD has evolved a unique strategy to evade host antileishmanial immune repertoire by manipulating host MyD88 to its advantage.
Collapse
Affiliation(s)
- Budhaditya Mukherjee
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Joydeep Paul
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Sandip Mukherjee
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Rupkatha Mukhopadhyay
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shantanabha Das
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Kshudiram Naskar
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Shyam Sundar
- Institute of Medical Sciences, Benaras Hindu University, Varanasi 221005, India
| | - Jean-Claude Dujardin
- Institute of Tropical Medicine, Antwerp 2000, Belgium; University of Antwerp, Antwerp 2000, Belgium; and
| | - Bhaskar Saha
- National Centre for Cell Science, Pune 411007, India
| | - Syamal Roy
- Department of Infectious Diseases and Immunology, Council of Scientific and Industrial Research, Indian Institute of Chemical Biology, Kolkata 700032, India;
| |
Collapse
|
19
|
Deconstructing innate immune signaling in myelodysplastic syndromes. Exp Hematol 2015; 43:587-598. [PMID: 26143580 DOI: 10.1016/j.exphem.2015.05.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 05/23/2015] [Indexed: 02/06/2023]
Abstract
Overexpression of immune-related genes is widely reported in myelodysplastic syndromes (MDSs), and chronic immune stimulation increases the risk for developing MDS. Aberrant innate immune activation, such as that caused by increased toll-like receptor (TLR) signaling, in MDS can contribute to systemic effects on hematopoiesis, in addition to cell-intrinsic defects on hematopoietic stem/progenitor cell (HSPC) function. This review will deconstruct aberrant function of TLR signaling mediators within MDS HSPCs that may contribute to cell-intrinsic consequences on hematopoiesis and disease pathogenesis. We will discuss the contribution of chronic TLR signaling to the pathogenesis of MDS based on evidence from patients and mouse genetic models.
Collapse
|
20
|
Zheng HY, Zheng HY, Zhou YT, Liu EL, Li J, Zhang YM. Changes of TIZ expression in epithelial ovarian cancer cells. ASIAN PAC J TROP MED 2015; 8:157-61. [PMID: 25902032 DOI: 10.1016/s1995-7645(14)60308-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 12/20/2014] [Accepted: 01/15/2015] [Indexed: 10/23/2022] Open
Abstract
OBJECTIVE To study the change of TIZ expression in epithelial ovarian cancer cells. METHODS HO8910 cells were transinfected with siRNA to inhibit the expression of TIZ. pcDNA3.1-TIZ vectors were combined to increase the TIZ expression level. The cell viability, colony forming efficiency and cycle distribution of HO8910, HO8910/NC, HO8910/pcDNA3.1-NC, HO8910/TIZ-573 and H08910/pcDNA3.1-TIZ were compared, and the invasion rate, migration rate and adhesion rate between 5 groups of cells were compared. RESULTS Compared with those of HO8910, HO8910/NC and HO8910/pcDNA3.1-NC, the cell viability, colony forming efficiency and cell cycle distribution of HO8910/TIZ-573 were increased, while the indexes of H08910/pcDNA3.1-NC were decreased with statistical significant difference (P<0.05). There was no statistical significant difference in the invasion rate, migration rate and adhesion rate between 5 groups of cells (P>0.05). CONCLUSIONS The expression of TIZ can inhibit the proliferation of epithelial ovarian cancer cells.
Collapse
Affiliation(s)
- Huan-Yu Zheng
- Department of Obstetrics & Gynecology, Affiliated Tangshan Workers Hospital of Hebei Medical University, Tangshan, Hebei 063000, China
| | - Hong-Yu Zheng
- Department of Pharmacy, Tangshan People's Hospital, Tangshan, Hebei 063000, China
| | - Yun-Tao Zhou
- Central Laboratory, Affiliated Tangshan Workers Hospital of Hebei Medical University, Tangshan, Hebei 063000, China
| | - En-Ling Liu
- Department of Obstetrics & Gynecology, Affiliated Tangshan Workers Hospital of Hebei Medical University, Tangshan, Hebei 063000, China.
| | - Jie Li
- Department of Obstetrics & Gynecology, Affiliated Tangshan Workers Hospital of Hebei Medical University, Tangshan, Hebei 063000, China
| | - Yan-Mei Zhang
- Department of Obstetrics & Gynecology, Affiliated Tangshan Workers Hospital of Hebei Medical University, Tangshan, Hebei 063000, China
| |
Collapse
|
21
|
Thaker YR, Schneider H, Rudd CE. TCR and CD28 activate the transcription factor NF-κB in T-cells via distinct adaptor signaling complexes. Immunol Lett 2014; 163:113-9. [PMID: 25455592 PMCID: PMC4286576 DOI: 10.1016/j.imlet.2014.10.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 07/28/2014] [Accepted: 10/15/2014] [Indexed: 01/07/2023]
Abstract
CD28 and TCR receptors use independent pathways to regulate NF-κB activation in T-cells. CD28 mediated NF-κB activation is dependent on the YMN-FM site for GRB-2 adaptor binding. The adaptors ADAP and SKAP1 are dispensable for direct CD28 activation of NF-κB. TCR driven NF-κB activation requires adaptor ADAP expression.
The transcription factor NF-κB is needed for the induction of inflammatory responses in T-cells. Whether its activation by the antigen-receptor and CD28 is mediated by the same or different intracellular signaling pathways has been unclear. Here, using T-cells from various knock-out (Cd28−/−, adap−/−) and knock-in (i.e. Cd28 Y-170F) mice in conjunction with transfected Jurkat T-cells, we show that the TCR and CD28 use distinct pathways to activate NF-κB in T-cells. Anti-CD28 ligation alone activated NF-κB in primary and Jurkat T-cells as measured by NF-κB reporter and EMSA assays. Anti-CD28 also activated NF-κB normally in primary T-cells from adap−/− mice, while anti-CD3 stimulation required the adaptor ADAP. Over-expression of ADAP or its binding partner SKAP1 failed to enhance anti-CD28 activation of NF-κB, while ADAP greatly increased anti-CD3 induced NF-κB activity. By contrast, CD28 activation of NF-κB depended on GRB-2 binding to CD28 as seen in CD28 deficient Jurkat T-cells reconstituted with the CD28 YMN-FM mutant, and in primary T-cells from CD28 Y170F mutant knock-in mice. CD28 associated with GRB-2, and GRB-2 siRNA impaired CD28 NF-κB activation. GRB-2 binding partner and guanine nucleotide exchange factor, VAV1, greatly enhanced anti-CD28 driven activation of NF-κB. Further, unlike in the case of anti-CD28, NF-κB activation by anti-CD3 and its cooperation with ADAP was strictly dependent on LAT expression. Overall, we provide evidence that CD28 and the TCR complex regulate NF-κB via different signaling modules of GRB-2/VAV1 and LAT/ADAP pathways respectively.
Collapse
Affiliation(s)
- Youg Raj Thaker
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom.
| | - Helga Schneider
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| | - Christopher E Rudd
- Cell Signalling Section, Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB2 1QP, United Kingdom
| |
Collapse
|
22
|
Mouse and human Notch-1 regulate mucosal immune responses. Mucosal Immunol 2014; 7:995-1005. [PMID: 24424521 DOI: 10.1038/mi.2013.118] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 11/22/2013] [Indexed: 02/04/2023]
Abstract
The Notch-1 signaling pathway is responsible for homeostatic tight junction expression in vitro, and promotes barrier function in vivo in the RAG1-adoptive transfer model of colitis. In this study, we sought to determine the role of colonic Notch-1 in the lymphoepithelial crosstalk in health and disease. We utilized in vivo and in vitro knockdown to target the expression of Notch-1. We identified that epithelial Notch-1 is required for appropriate activation of intestinal epithelial cells at steady state and upon inflammatory stimulus. Notch-1 expression modulates mucosal chemokine and cytokine secretion, and FoxP3 and effector T-cell responses. We showed that epithelial Notch-1 controls the immune function of the epithelium through crosstalk with the nuclear factor-κB (NF-κB)/mitogen-activated protein kinase (MAPK) pathways that, in turn, elicits T-cell responses. Overall, epithelial Notch-1 bridges innate and adaptive immunity in the gut. Our findings highlight an indispensable role for Notch-1-mediated signaling in the intricate epithelial-immune crosstalk, and validate that epithelial Notch-1 is necessary and sufficient to support protective epithelial proinflammatory responses.
Collapse
|
23
|
Askarian F, van Sorge NM, Sangvik M, Beasley FC, Henriksen JR, Sollid JUE, van Strijp JAG, Nizet V, Johannessen M. A Staphylococcus aureus TIR domain protein virulence factor blocks TLR2-mediated NF-κB signaling. J Innate Immun 2014; 6:485-98. [PMID: 24481289 DOI: 10.1159/000357618] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 11/25/2013] [Indexed: 01/05/2023] Open
Abstract
Signaling through Toll-like receptors (TLRs), crucial molecules in the induction of host defense responses, requires adaptor proteins that contain a Toll/interleukin-1 receptor (TIR) domain. The pathogen Staphylococcus aureus produces several innate immune-evasion molecules that interfere with the host's innate immune response. A database search analysis suggested the presence of a gene encoding a homologue of the human TIR domain in S. aureus MSSA476 which was named staphylococcal TIR domain protein (TirS). Ectopic expression of TirS in human embryonic kidney, macrophage and keratinocyte cell lines interfered with signaling through TLR2, including MyD88 and TIRAP, NF-κB and/or mitogen-activated protein kinase pathways. Moreover, the presence of TirS reduced the levels of cytokines MCP-1 and G-CSF secreted in response to S. aureus. The effects on NF-κB pathway were confirmed using S. aureus MSSA476 wild type, an isogenic mutant MSSA476ΔtirS, and complemented MSSA476ΔtirS +pTirS in a Transwell system where bacteria and host cells were physically separated. Finally, in a systematic mouse infection model, TirS promoted bacterial accumulation in several organs 4 days postinfection. The results of this study reveal a new S. aureus virulence factor that can interfere with PAMP-induced innate immune signaling in vitro and bacterial survival in vivo.
Collapse
Affiliation(s)
- Fatemeh Askarian
- Research Group of Host-Microbe Interactions, Department of Medical Biology, Faculty of Health Sciences, UiT The Artic University of Norway, Tromsø, Norway
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Interleukin-1-induced activation of the small GTPase Rac1 depends on receptor internalization and regulates gene expression. Cell Signal 2014; 26:49-55. [DOI: 10.1016/j.cellsig.2013.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 09/19/2013] [Accepted: 09/19/2013] [Indexed: 11/19/2022]
|
25
|
Xiao L, Liu Y, Wang N. New paradigms in inflammatory signaling in vascular endothelial cells. Am J Physiol Heart Circ Physiol 2013; 306:H317-25. [PMID: 24285111 DOI: 10.1152/ajpheart.00182.2013] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Inflammation is a basic cellular process in innate and adaptive immunity. Vascular endothelial cells play an important role in the initiation, amplification, and resolution of the inflammatory response. Deregulated inflammatory response is implicated in a variety of cardiovascular diseases such as atherosclerosis, obesity, diabetes, and hypertension. Recent studies have made significant progresses in the understanding of the complex molecular pathways that mediate the pro- and anti-inflammatory signaling in endothelial cells (ECs). Specifically, a number of macromolecular complexes termed as signalosomes have been identified to integrate the proinflammatory signaling from the membrane receptors to key transcription factors such as nuclear factor-κB (NF-κB). Inflammasomes are associated with the pattern-recognition receptors such as Toll-like receptors (TLRs), nucleotide-binding oligomerization-domain (NOD)-like receptors (NLRs) to mediate innate immunity responses. Emerging evidence has also revealed that noncoding microRNAs constitute a new class of intra- and intercellular signaling molecules to modulate inflammation in ECs. Thus this article will briefly summarize these new mechanisms with a special emphasis in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Lei Xiao
- Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | | | | |
Collapse
|
26
|
Hamilton T, Li X, Novotny M, Pavicic PG, Datta S, Zhao C, Hartupee J, Sun D. Cell type- and stimulus-specific mechanisms for post-transcriptional control of neutrophil chemokine gene expression. J Leukoc Biol 2011; 91:377-83. [PMID: 22167720 DOI: 10.1189/jlb.0811404] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
mRNAs encoding inflammatory chemokines that recruit neutrophils frequently exhibit short half-lives that serve to limit their expression under inappropriate conditions but are often prolonged to ensure adequate levels during inflammatory response. Extracellular stimuli that modulate the stability of such mRNAs may be the same as the transcriptional activator, as is the case with TLR ligands, or may cooperate with independent transcriptional stimuli, as with IL-17, which extends the half-life of TNF-induced transcripts. These different stimuli engage independent signaling pathways that target different instability mechanisms distinguished by dependence on different regulatory nucleotide sequence motifs within the 3'UTRs, which involve that action of different mRNA-binding proteins. The selective use of these pathways by different stimuli and in distinct cell populations provides the potential for tailoring of chemokine expression patterns to meet specific needs in different pathophysiologic circumstances.
Collapse
Affiliation(s)
- Thomas Hamilton
- Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195-0001, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Rebl A, Rebl H, Liu S, Goldammer T, Seyfert HM. Salmonid Tollip and MyD88 factors can functionally replace their mammalian orthologues in TLR-mediated trout SAA promoter activation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2011; 35:81-87. [PMID: 20813127 DOI: 10.1016/j.dci.2010.08.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 08/23/2010] [Accepted: 08/24/2010] [Indexed: 05/29/2023]
Abstract
Many functional details of the piscine Toll-like receptor (TLR) signal-mediated activation of immune defense are still elusive. We used an established reconstitution system of mammalian TLR signaling to examine if this system would allow for pathogen-dependent promoter activation of the serum amyloid A (SAA)-encoding gene from rainbow trout (Oncorhynchus mykiss) and if the key mediators MyD88 and Tollip from trout can functionally substitute for their mammalian orthologues. Cells of the established human embryonic kidney line HEK-293 were transiently co-transfected with vectors expressing bovine TLR2 or TLR4 factors and a reporter gene driven by the promoter of the trout SAA gene. Escherichia coli stimulation increased reporter gene expression more than 3-fold. Deletion series and point mutations identified in the proximal SAA promoter a composite overlapping binding site for NF-κB and CEBP factors as crucial for promoter activation. Overexpression of NF-κB p65, but not of p50 or different members of the CEBP factor family proved this factor as an essential driver for SAA expression. Overexpression of a transdominant-negative mutant of the trout MyD88 factor reduced TLR-mediated SAA promoter activation confirming functional conservation of its TIR domain. Overexpression of the Tollip factor from trout also quenched TLR-mediated NF-κB and TLR4-mediated SAA promoter activation. The MyD88 mutant and Tollip expression studies confirm the functional homology of both piscine factors and their mammalian counterparts. We provide for the first time evidence that also the Tollip-mediated negative loop of TLR signaling may be conserved in non-mammalian organisms.
Collapse
Affiliation(s)
- Alexander Rebl
- Leibniz Institute for Farm Animal Biology (FBN), Molecular Biology Research Unit, Wilhelm-Stahl-Allee 2, 18196 Dummerstorf, Germany
| | | | | | | | | |
Collapse
|
28
|
Fernandes A, Barateiro A, Falcão AS, Silva SLA, Vaz AR, Brito MA, Silva RFM, Brites D. Astrocyte reactivity to unconjugated bilirubin requires TNF-α and IL-1β receptor signaling pathways. Glia 2010; 59:14-25. [PMID: 20967881 DOI: 10.1002/glia.21072] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 07/30/2010] [Accepted: 08/10/2010] [Indexed: 12/31/2022]
Abstract
Jaundice and sepsis are common neonatal conditions that can lead to neurodevelopment sequelae, namely if present at the same time. We have reported that tumor necrosis factor (TNF)-α and interleukin (IL)-1β are produced by cultured neurons and mainly by glial cells exposed to unconjugated bilirubin (UCB). The effects of these cytokines are mediated by cell surface receptors through a nuclear factor (NF)-κB-dependent pathway that we have showed to be activated by UCB. The present study was designed to evaluate the role of TNF-α and IL-1β signaling on astrocyte reactivity to UCB in rat cortical astrocytes. Exposure of astrocytes to UCB increased the expression of both TNF-α receptor (TNFR)1 and IL-1β receptor (IL-1R)1, but not TNFR2, as well as their activation, observed by augmented binding of receptors' molecular adaptors, TRAF2 and TRAF6, respectively. Silencing of TNFR1, using siRNA technology, or blockade of IL-1β cascade, using its endogenous antagonist, IL-1 receptor antagonist (IL-1ra), prevented UCB-induced cytokine release and NF-κB activation. Interestingly, lack of TNF-α signal transduction reduced UCB-induced cell death for short periods of incubation, although an increase was observed after extended exposure; in contrast, inhibition of IL-1β cascade produced a sustained blockade of astrocyte injury by UCB. Together, our data show that inflammatory pathways are activated during in vitro exposure of rat cortical astrocytes to UCB and that this activation is prolonged in time. This supports the concept that inflammatory pathways play a role in brain damage by UCB, and that they may represent important pharmacological targets.
Collapse
Affiliation(s)
- Adelaide Fernandes
- Research Institute for Medicines and Pharmaceutical Sciences (IMedUL), Faculdade de Farmácia, University of Lisbon, Lisbon, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Rosales-Corral S, Reiter RJ, Tan DX, Ortiz GG, Lopez-Armas G. Functional aspects of redox control during neuroinflammation. Antioxid Redox Signal 2010; 13:193-247. [PMID: 19951033 DOI: 10.1089/ars.2009.2629] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neuroinflammation is a CNS reaction to injury in which some severe pathologies, regardless of their origin, converge. The phenomenon emphasizes crosstalk between neurons and glia and reveals a complex interaction with oxidizing agents through redox sensors localized in enzymes, receptors, and transcription factors. When oxidizing pressures cause reversible molecular changes, such as minimal or transitory proinflammatory cytokine overproduction, redox couples provide a means of translating the presence of reactive oxygen or nitrogen species into useful signals in the cell. Additionally, thiol-based redox sensors convey information about localized changes in redox potential induced by physiologic or pathologic situations. They are susceptible to oxidative changes and become key events during neuroinflammation, altering the course of a signaling response or the behavior of specific transcription factors. When oxidative stress augments the pressure on the intracellular environment, the effective reduction potential of redox pairs diminishes, and cell signaling shifts toward proinflammatory and proapoptotic signals, creating a vicious cycle between oxidative stress and neuroinflammation. In addition, electrophilic compounds derived from the oxidative cascade react with key protein thiols and interfere with redox signaling. This article reviews the relevant functional aspects of redox control during the neuroinflammatory process.
Collapse
Affiliation(s)
- Sergio Rosales-Corral
- Lab. Desarrollo-Envejecimiento, Enfermedades Neurodegenerativas, División de Neurociencias, Centro de Investigación Biomédica de Occidente (CIBO) del Instituto Mexicano del Seguro Social (IMSS) , Guadalajara, Jalisco. Mexico.
| | | | | | | | | |
Collapse
|
30
|
Hamilton T, Novotny M, Pavicic PJ, Herjan T, Hartupee J, Sun D, Zhao C, Datta S. Diversity in post-transcriptional control of neutrophil chemoattractant cytokine gene expression. Cytokine 2010; 52:116-22. [PMID: 20430641 DOI: 10.1016/j.cyto.2010.04.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Accepted: 04/05/2010] [Indexed: 12/11/2022]
Abstract
Regulation of neutrophil chemokine gene expression represents an important feature in tissue inflammation. While chemokine gene transcription through the action of NFkappaB is recognized as an essential component of this process, it is now clear that post-transcriptional mechanisms, particularly the rates of decay of mature cytoplasmic mRNA, provides an essential component of this control. Chemokine and other cytokine mRNA half life is known to be controlled via adenine-uridine rich sequence motifs localized within 3' untranslated regions (UTRs), the most common of which contains one or more copies of the pentameric AUUUA sequence. In myeloid cells AUUUA sequences confer instability through the action of RNA binding proteins such as tristetraprolin (TTP). The resulting instability can be regulated in response to extra-cellular stimuli including Toll like receptor ligands that signal to control the function of TTP through pathways involving the activation of p38 MAP kinases. Recent findings indicate that substantial mechanistic diversity is operative in non-myeloid cells in response to alternate pro-inflammatory stimuli such as IL-17. These pathways target distinct instability sequences that do not contain the AUUUA pentamer motif, do not signal through p38 MAPK, and function independently of TTP.
Collapse
Affiliation(s)
- Thomas Hamilton
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Maitra U, Singh N, Gan L, Ringwood L, Li L. IRAK-1 contributes to lipopolysaccharide-induced reactive oxygen species generation in macrophages by inducing NOX-1 transcription and Rac1 activation and suppressing the expression of antioxidative enzymes. J Biol Chem 2010; 284:35403-11. [PMID: 19850916 DOI: 10.1074/jbc.m109.059501] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Inflammatory stimulants such as bacterial endotoxin (lipopolysaccharide (LPS)) are known to induce tissue damage and injury partly through the induction of reactive oxygen species (ROS). Although it is recognized that the induction of ROS in macrophages by LPS depends upon the expression and activation of NADPH oxidase, as well as the suppression of antioxidative enzymes involved in ROS clearance, the underlying molecular mechanisms are poorly defined. In this study, we examined the contribution of the interleukin-1 receptor-associated kinase 1 (IRAK-1) to LPS-induced generation of ROS. We observed that LPS induced significantly less ROS in IRAK-1(-/-) macrophages, indicating that IRAK-1 is critically involved in the induction of ROS. Mechanistically, we observed that IRAK-1 is required for LPS-induced expression of NOX-1, a key component of NADPH oxidase, via multiple transcription factors, including p65/RelA, C/EBPbeta, and C/EBPdelta. On the other hand, we demonstrated that IRAK-1 associated with and activated small GTPase Rac1, a known activator of NOX-1 oxidase enzymatic activity. IRAK-1 forms a close complex with Rac1 via a novel LWPPPP motif within the variable region of IRAK-1. On the other hand, we also observed that IRAK-1 is required for LPS-mediated suppression of peroxisome proliferator-activated receptor alpha and PGC-1alpha, nuclear factors essential for the expression of antioxidative enzymes such as GPX3 and catalase. Consequently, injection of LPS causes significantly less plasma lipid peroxidation in IRAK-1(-/-) mice compared with wild type mice. Taken together, our study reveals IRAK-1 as a novel component involved in the generation of ROS induced by LPS.
Collapse
Affiliation(s)
- Urmila Maitra
- Departments of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, USA
| | | | | | | | | |
Collapse
|
32
|
Santos-Sierra S, Deshmukh SD, Kalnitski J, Küenzi P, Wymann MP, Golenbock DT, Henneke P. Mal connects TLR2 to PI3Kinase activation and phagocyte polarization. EMBO J 2009; 28:2018-27. [PMID: 19574958 PMCID: PMC2718282 DOI: 10.1038/emboj.2009.158] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2008] [Accepted: 05/22/2009] [Indexed: 02/06/2023] Open
Abstract
The recognition of bacterial lipoproteins by toll-like receptor (TLR) 2 is pivotal for inflammation initiation and control in many bacterial infections. TLR2-dependent signalling is currently believed to essentially require both adaptor proteins MyD88 (myeloid differentiation primary response gene 88) and Mal/TIRAP (MyD88-adapter-like/TIR-domain-containing adaptor protein). TLR2-dependent, but MyD88-independent responses have not been described yet. We report here on a novel-signalling pathway downstream of TLR2, which does not adhere to the established model. On stimulation of the TLR2/6 heterodimer with diacylated bacterial lipoproteins, Mal directly interacts with the regulatory subunit of phosphoinositide 3-kinase (PI3K), p85alpha, in an inducible fashion. The Mal-p85alpha interaction drives PI3K-dependent phosphorylation of Akt, phosphatidylinositol(3,4,5)P3 (PIP(3)) generation and macrophage polarization. MyD88 is not essential for PI3K activation and Akt phosphorylation; however, cooperates with Mal for PIP(3) formation and accumulation at the leading edge. In contrast to TLR2/6, TLR2/1 does not require Mal or MyD88 for Akt phosphorylation. Hence, Mal specifically connects TLR2/6 to PI3K activation, PIP(3) generation and macrophage polarization.
Collapse
Affiliation(s)
- S Santos-Sierra
- Center for Pediatrics and Adolescent Medicine and Centre of Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - S D Deshmukh
- Center for Pediatrics and Adolescent Medicine and Centre of Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - J Kalnitski
- Center for Pediatrics and Adolescent Medicine and Centre of Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - P Küenzi
- Institute of Pharmaceutical Biology, University of Basel, Basel, Switzerland
| | - M P Wymann
- Institute of Biochemistry and Genetics, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - D T Golenbock
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Philipp Henneke
- Center for Pediatrics and Adolescent Medicine and Centre of Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Oakley FD, Abbott D, Li Q, Engelhardt JF. Signaling components of redox active endosomes: the redoxosomes. Antioxid Redox Signal 2009; 11:1313-33. [PMID: 19072143 PMCID: PMC2842130 DOI: 10.1089/ars.2008.2363] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Subcellular compartmentalization of reactive oxygen species (ROS) plays a critical role in transmitting cell signals in response to environmental stimuli. In this regard, signals at the plasma membrane have been shown to trigger NADPH oxidase-dependent ROS production within the endosomal compartment and this step can be required for redox-dependent signal transduction. Unique features of redox-active signaling endosomes can include NADPH oxidase complex components (Nox1, Noxo1, Noxa1, Nox2, p47phox, p67phox, and/or Rac1), ROS processing enzymes (SOD1 and/or peroxiredoxins), chloride channels capable of mediating superoxide transport and/or membrane gradients required for Nox activity, and novel redox-dependent sensors that control Nox activity. This review will discuss the cytokine and growth factor receptors that likely mediate signaling through redox-active endosomes, and the common mechanisms whereby they act. Additionally, the review will cover ligand-independent environmental injuries, such as hypoxia/reoxygenation injury, that also appear to facilitate cell signaling through NADPH oxidase at the level of the endosome. We suggest that redox-active endosomes encompass a subset of signaling endosomes that we have termed redoxosomes. Redoxosomes are uniquely equipped with redox-processing proteins capable of transmitting ROS signals from the endosome interior to redox-sensitive effectors on the endosomal surface. In this manner, redoxosomes can control redox-dependent effector functions through the spatial and temporal regulation of ROS as second messengers.
Collapse
Affiliation(s)
- Fredrick D Oakley
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | |
Collapse
|
34
|
Rac1 regulates peptidoglycan-induced nuclear factor-κB activation and cyclooxygenase-2 expression in RAW 264.7 macrophages by activating the phosphatidylinositol 3-kinase/Akt pathway. Mol Immunol 2009; 46:1179-88. [DOI: 10.1016/j.molimm.2008.11.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2006] [Revised: 11/10/2008] [Accepted: 11/11/2008] [Indexed: 01/22/2023]
|
35
|
Lee NK, Choi HK, Kim DK, Lee SY. Rac1 GTPase regulates osteoclast differentiation through TRANCE-induced NF-kappa B activation. Mol Cell Biochem 2009; 281:55-61. [PMID: 16328957 DOI: 10.1007/s11010-006-0333-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Accepted: 06/27/2005] [Indexed: 10/25/2022]
Abstract
Signaling by tumor necrosis factor (TNF)-related activation-induced cytokine (TRANCE) is essential for the differentiation of monocytes/macrophages into osteoclasts. We show here that TRANCE selectively activates Rac1, but not Rac2 in osteoclast precursors. Expression of a dominant interfering mutant of TNF receptor-associated factor (TRAF)6 blocks TRANCE-mediated Rac1 activation, indicating that Rac1 lies downstream of TRAF6. Osteoclast precursors expressing a dominant negative Rac1N17 are defective in TRANCE-induced IKK activation and IkappaBalpha degradation resulting in inhibition of NFkappaB-dependent reporter gene activity. In addition, Rac1 acts upstream of TAK1 to induce NF-kappaB activation and is required for the normal differentiation of osteoclast precursors. Thus, Rac1 may represent a key regulator for differentiation of osteoclasts through the activation of NF-kappaB.
Collapse
Affiliation(s)
- Na Kyung Lee
- Division of Molecular Life Sciences and Center for Cell Signaling Research, Ewha Womans University, Seoul 120-750, Korea.
| | | | | | | |
Collapse
|
36
|
Stack J, Bowie AG. Characterisation of viral proteins that inhibit Toll-like receptor signal transduction. Methods Mol Biol 2009; 517:217-235. [PMID: 19378031 DOI: 10.1007/978-1-59745-541-1_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Toll-like receptor (TLR) signalling involves five TIR adapter proteins, which couple to downstream protein kinases that ultimately lead to the activation of transcription factors such as nuclear factor-kappaB (NF-kappaB) and members of the interferon regulatory factor (IRF) family. TLRs play a crucial role in host defence against invading microorganisms, and highlighting their importance in the immune system is the fact that TLRs are targeted by viral immune evasion strategies. Identifying the target host proteins of such viral inhibitors is very important because valuable insights into how host cells respond to infection may be obtained. Also, viral proteins may have potential as therapeutic agents. Luciferase reporter gene assays are a very useful tool for the analysis of TLR signalling pathways, as the effect of a putative viral inhibitor on a large amount of signals can be examined in one experiment. A basic reporter gene assay involves the transfection of cells with a luciferase reporter gene, along with an activating expression plasmid, with or without a plasmid expressing a viral inhibitor. Induction of a signalling pathway leads to luciferase protein expression, which is measured using a luminometer. Results from these assays can be informative for deciding which host proteins to test for interactions with a viral inhibitor. Successful assays for measuring protein-protein interactions include co-immunoprecipitations (Co-IPs) and glutathione-S-transferase (GST)-pulldowns. Co-IP experiments involve precipitating a protein out of a cell lysate using a specific antibody bound to Protein A/G sepharose. Additional molecules complexed to that protein are captured as well and can be detected by Western blot analysis. GST-pulldown experiments are similar in principle to Co-IPs, but a bait GST-fusion protein complexed to glutathione-sepharose (GSH) beads is used to pull down interaction partners instead of an antibody. Again, complexes recovered from the beads are analysed by Western blotting.
Collapse
Affiliation(s)
- Julianne Stack
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin 2, Ireland
| | | |
Collapse
|
37
|
Lee Y, Kim H, Kim S, Shin MH, Kim YK, Kim KH, Chung JH. Myeloid differentiation factor 88 regulates basal and UV-induced expressions of IL-6 and MMP-1 in human epidermal keratinocytes. J Invest Dermatol 2008; 129:460-7. [PMID: 18719610 DOI: 10.1038/jid.2008.261] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myeloid differentiation factor 88 (MyD88) is known as an adaptor protein for the Toll-like receptor (TLR) family and participates in signal transduction by binding to the cytoplasmic Toll/IL-1 receptor (TIR) domains of activated TLR. In this study, we demonstrated that expression of MyD88 is increased in photoaged skin compared with intrinsic aged human skin of the same elderly individuals, and that acute UV irradiation increases MyD88 expression in human skin in vivo. To investigate the effects of these high levels of MyD88 in photoaged skin and acutely UV-irradiated skin, human epidermal keratinocytes were infected with adenovirus expressing wild-type (MyD88wt), dominant-positive (MyD88DeltaC), and dominant-negative (MyD88DeltaN) MyD88 forms. Overexpression of MyD88wt and MyD88DeltaC, but not of MyD88DeltaN, increased the basal expressions of IL-6 and matrix metalloproteinase-1 (MMP-1) in human epidermal keratinocytes. Moreover, overexpression of MyD88DeltaN prevented UV-induced expressions of IL-6 and MMP-1 by inhibiting UV-induced activation of NF-kappaB and activating protein-1. These results suggest that MyD88 is important in IL-6 and MMP-1 expressions in both acutely UV-irradiated skin and in chronically sun-exposed human skin.
Collapse
Affiliation(s)
- Youngae Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
38
|
Bao X, Sinha M, Liu T, Hong C, Luxon B, Garofalo R, Casola A. Identification of human metapneumovirus-induced gene networks in airway epithelial cells by microarray analysis. Virology 2008; 374:114-27. [PMID: 18234263 PMCID: PMC2777699 DOI: 10.1016/j.virol.2007.12.024] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 10/17/2007] [Accepted: 12/17/2007] [Indexed: 01/21/2023]
Abstract
Human metapneumovirus (hMPV) is a major cause of lower respiratory tract infections in infants, elderly and immunocompromised patients. Little is known about the response to hMPV infection of airway epithelial cells, which play a pivotal role in initiating and shaping innate and adaptive immune responses. In this study, we analyzed the transcriptional profiles of airway epithelial cells infected with hMPV using high-density oligonucleotide microarrays. Of the 47,400 transcripts and variants represented on the Affimetrix GeneChip Human Genome HG-U133 plus 2 array, 1601 genes were significantly altered following hMPV infection. Altered genes were then assigned to functional categories and mapped to signaling pathways. Many up-regulated genes are involved in the initiation of pro-inflammatory and antiviral immune responses, including chemokines, cytokines, type I interferon and interferon-inducible proteins. Other important functional classes up-regulated by hMPV infection include cellular signaling, gene transcription and apoptosis. Notably, genes associated with antioxidant and membrane transport activity, several metabolic pathways and cell proliferation were down-regulated in response to hMPV infection. Real-time PCR and Western blot assays were used to confirm the expression of genes related to several of these functional groups. The overall result of this study provides novel information on host gene expression upon infection with hMPV and also serves as a foundation for future investigations of genes and pathways involved in the pathogenesis of this important viral infection. Furthermore, it can facilitate a comparative analysis of other paramyxoviral infections to determine the transcriptional changes that are conserved versus the one that are specific to individual pathogens.
Collapse
Affiliation(s)
- X. Bao
- Department of Pediatrics, University of Texas Medical Branch, Galveston Texas
| | - M. Sinha
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston Texas
- UTMB Bioinformatics Program, University of Texas Medical Branch, Galveston Texas
| | - T. Liu
- Department of Pediatrics, University of Texas Medical Branch, Galveston Texas
| | - C. Hong
- Department of Pediatrics, University of Texas Medical Branch, Galveston Texas
| | - B.A Luxon
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston Texas
- UTMB Bioinformatics Program, University of Texas Medical Branch, Galveston Texas
| | - R.P. Garofalo
- Department of Pediatrics, University of Texas Medical Branch, Galveston Texas
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston Texas
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston Texas
| | - A. Casola
- Department of Pediatrics, University of Texas Medical Branch, Galveston Texas
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston Texas
- Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston Texas
| |
Collapse
|
39
|
Interleukin-1 stimulates glutamate uptake in glial cells by accelerating membrane trafficking of Na+/K+-ATPase via actin depolymerization. Mol Cell Biol 2008; 28:3273-80. [PMID: 18332114 DOI: 10.1128/mcb.02159-07] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Interleukin-1 (IL-1) is a mediator of brain injury induced by ischemia, trauma, and chronic neurodegenerative disease. IL-1 also has a protective role by preventing neuronal cell death from glutamate neurotoxicity. However, the cellular mechanisms of IL-1 action remain unresolved. In the mammalian retina, glutamate/aspartate transporter (GLAST) is a Na(+)-dependent, major glutamate transporter localized to Müller glial cells, and loss of GLAST leads to glaucomatous retinal degeneration (T. Harada, C. Harada, K. Nakamura, H. A. Quah, A. Okumura, K. Namekata, T. Saeki, M. Aihara, H. Yoshida, A. Mitani, and K. Tanaka, J. Clin. Investig. 117:1763-1770, 2007). We show here that IL-1 increases glutamate uptake in Müller cells by a mechanism that involves increased membrane Na(+)/K(+)-ATPase localization, required for counteracting the Na(+)-glutamate cotransport. IL-1 activated the p38 mitogen-activated protein kinase (MAPK)/capase 11 pathway, which destabilizes the actin cytoskeleton allowing Na(+)/K(+)-ATPase membrane redistribution. Furthermore, pretreatment with IL-1 protected retinal neurons from glutamate neurotoxicity through p38 MAPK signaling. Our observations suggested that IL-1 acts as a potential neuroprotective agent by modulating the functions of the glia-neuron network.
Collapse
|
40
|
Konat GW, Krasowska-Zoladek A, Kraszpulski M. Statins enhance toll-like receptor 4-mediated cytokine gene expression in astrocytes: Implication of Rho proteins in negative feedback regulation. J Neurosci Res 2008; 86:603-9. [PMID: 17896797 DOI: 10.1002/jnr.21509] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Toll-like receptors (TLRs) are sentinels of innate immunity that recognize pathogenic molecules and trigger inflammatory response. Because inflammatory mediators are detrimental to the host, the TLR response is regulated by feedback inhibition. Statins, the inhibitors of isoprenoid biosynthesis, have been shown to be potent modulators of TLR activity, and this modulation may provide insight regarding mechanisms of the feedback inhibition. In the present study, we examined feedback mechanisms that regulate TLR4 activity in astrocytes using statins to perturb postligational signaling. Astrocytic cultures established from newborn rat brains were exposed to lipopolysaccharide (LPS), the ligand for TLR4. The up-regulation of expression of genes encoding interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha (TNFalpha) was determined by real-time RT-PCR. Pretreatment of the cells with either atorvastatin or simvastatin enhanced the LPS-induced up-regulation of cytokine gene expression. The most profound enhancement of approximately 17-fold was observed for the Il-6 gene. The enhancements for the Tnfa and Il-1b genes were approximately 5- and 3.5-fold, respectively. Mevalonate fully reversed the effects of statins, indicating that these drugs act through the inhibition of isoprenoid synthesis. The inhibition of protein geranylgeranylation, but not protein farnesylation, mimicked the effects of statins, strongly indicating that the enhancement is mediated by the Rho proteins. In support of this notion, pretreatment of cells with toxin B, a specific inhibitor of the Rho proteins, also enhanced LPS-triggered up-regulation of the cytokine genes. These results indicate that the Rho proteins are involved in the activation of negative feedback inhibition of TLR4 signaling in astrocytes.
Collapse
Affiliation(s)
- Gregory W Konat
- Department of Neurobiology and Anatomy, West Virginia University School of Medicine, Morgantown, West Virginia 26506-9128, USA.
| | | | | |
Collapse
|
41
|
Liu G, Tsuruta Y, Gao Z, Park YJ, Abraham E. Variant IL-1 receptor-associated kinase-1 mediates increased NF-kappa B activity. THE JOURNAL OF IMMUNOLOGY 2007; 179:4125-34. [PMID: 17785851 DOI: 10.4049/jimmunol.179.6.4125] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-1R-associated kinase (IRAK)-1 is a critical mediator of TLR/IL-1R-induced activation of the transcription factor NF-kappaB. We previously described that a commonly occurring IRAK-1 variant haplotype, containing amino acid changes from serine to phenylalanine at position 196 and from leucine to serine at position 532, is associated with increased activation of NF-kappaB in LPS-stimulated neutrophils from patients with sepsis-induced acute lung injury and also higher mortality and more severe clinical outcomes in such patients. To investigate the underlying molecular mechanisms, we examined the ability of wild-type and variant IRAK-1 to modulate NF-kappaB activation. We found increased NF-kappaB transcriptional activity and expression of NF-kappaB-dependent proinflammatory cytokines in IL-1beta-stimulated IRAK-1-deficient cells transfected with variant IRAK-1 as compared with IRAK-1 wild type. IkappaB-alpha degradation was faster and p65 phosphorylation more prolonged after IL-1beta stimulation in cells expressing the IRAK-1 variant. However, IL-1-induced activation of MAPKs and nuclear translocation of NF-kappaB are comparable in both IRAK-1 variant- and IRAK-1 wild-type-expressing cells. Autophosphorylation of the IRAK-1 variant is greater than that found with wild-type IRAK-1. Additionally, variant IRAK-1 has greater interaction with TNFR-associated factor 6 than does wild-type IRAK-1. The enhanced activity of variant IRAK-1 appeared to be due to the alteration at aa 532, with only minimal effects being associated with change at aa 196. These results demonstrate that variant IRAK-1 is associated with alterations in multiple intracellular events that are likely to contribute to increased NF-kappaB activation and inflammatory responses in individuals with this IRAK-1 haplotype.
Collapse
Affiliation(s)
- Gang Liu
- Department of Medicine, University of Alabama, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
42
|
Mizobe T, Tsukada J, Higashi T, Mouri F, Matsuura A, Tanikawa R, Minami Y, Yoshida Y, Tanaka Y. Constitutive association of MyD88 to IRAK in HTLV-I-transformed T cells. Exp Hematol 2007; 35:1812-22. [PMID: 17920759 DOI: 10.1016/j.exphem.2007.08.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2006] [Revised: 08/06/2007] [Accepted: 08/07/2007] [Indexed: 11/18/2022]
Abstract
OBJECTIVE Constitutive activation of nuclear factor (NF)-kappaB is a common feature of human T-cell leukemia virus type I (HTLV-I)-transformed T cells. Inhibition of NF-kappaB activity reduces cell growth and induces apoptosis of HTLV-I-transformed T cells, suggesting a central role of NF-kappaB in their proliferation and survival. In this study, we investigated whether MyD88, an adaptor protein of Toll-like receptor (TLR) signaling, contributes to constitutive NF-kappaB activation in HTLV-I-transformed T cells. MATERIALS AND METHODS Activation status of MyD88 and interleukin (IL)-1R-associated kinase 1 (IRAK1) in HTLV-I-transformed human T cells, MT2, MT4, and HUT102 was examined by using Western blot and immunoprecipitation. TLR expression was evaluated with reverse transcription polymerase chain reaction. An expression vector encoding a dominant negative MyD88 with a deletion of its death domain (MyD88dn) was transfected into MT2 cells to evaluate roles of MyD88 in spontaneous activation of cytokine gene promoters and transcription factors, proliferation, and apoptosis in HTLV-I-transformed T cells. RESULTS Constitutive association of MyD88 with IRAK1 was observed in all three of HTLV-I-transformed T cells, but not in HTLV-I-negative T cells, such as Jurkat, HUT78, and MOLT4. MT2 cells showed expression of TLR-1, -6, and -10 mRNAs. Constitutive activation of NF-kappaB and NF-IL-6 and cytokine gene promoters, such as IL-1alpha, interferon-gamma, and tumor necrosis factor-alpha in MT2 cells was inhibited by MyD88dn expression. MyD88dn reduced proliferation and induced apoptosis of MT2 cells. HTLV-I Tax enhanced TLR expression and synergistically activated NF-kappaB with wild-type MyD88. CONCLUSION Our results show a novel pathway in NF-kappaB activation in HTLV-I-transformed T cells and further demonstrate a critical role of MyD88 in their dysregulated gene activation, survival, and proliferation.
Collapse
Affiliation(s)
- Takamitsu Mizobe
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sakoda K, Yamamoto M, Negishi Y, Liao JK, Node K, Izumi Y. Anti-inflammatory Effects of Simvastatin on Human Oral Cells. Inflamm Regen 2007. [DOI: 10.2492/inflammregen.27.107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
44
|
Steinberg T, Dannewitz B, Tomakidi P, Hoheisel JD, Müssig E, Kohl A, Nees M. Analysis of interleukin-1β-modulated mRNA gene transcription in human gingival keratinocytes by epithelia-specific cDNA microarrays. J Periodontal Res 2006; 41:426-46. [PMID: 16953820 DOI: 10.1111/j.1600-0765.2006.00884.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND/OBJECTIVES Proinflammatory cytokines such as interleukin-1beta are known to be synthesized in oral gingivitis and periodontitis and lead to the activation of the transcription factor nuclear factor-kappaB (NF-kappaB). Although numerous effects of interleukin-1beta on mesenchymal cells are known, e.g. up-regulation of intercellular adhesion molecule-1 in endothelial cells, little is known of the effects of interleukin-1beta on oral keratinocytes. The purpose of the present study was to seek interleukin-1beta-mediated alterations in mRNA gene transcription and a putative activation of NF-kappaB in oral gingival keratinocytes. METHODS As an in vitro model for gingivitis and periodontitis, immortalized human gingival keratinocytes (IHGK) were stimulated with the proinflammatory cytokine interleukin-1beta. An epithelia-specific cDNA microarray was used to analyze mRNA expression profiles from IHGK cells treated with 200 units interleukin-1beta/ml for 3, 6, 9, 12, and 24 h. Indirect immunofluorescence was carried out to detect NF-kappaB in IHGK following interleukin-1beta treatment. RESULTS Detailed analysis revealed distinct patterns of time-dependent changes, including genes induced or repressed early (3-6 h) or late (12-24 h) after interleukin-1beta treatment. Differentially expressed genes were involved in (i) cell stress, (ii) DNA repair, (iii) cell cycle and proliferation, (iv) anti-pathogen response, (v) extracellular matrix turnover, and (vi) angiogenesis. A large number of genes were responsive to NF-kappaB and induction was concomitant with nuclear translocation of the p65 RelA subunit of NF-kappaB. Interestingly, many of these genes contain multiple NF-kappaB binding sites in their promoters. CONCLUSION Analysis of altered gene expression allows identification of gene networks associated with inflammatory responses. In addition to a number of well-known genes involved in gingivitis and periodontitis, we identified novel candidates that might be associated with the onset and maintenance of an inflammatory disease.
Collapse
Affiliation(s)
- T Steinberg
- Department of Orthodontics and Dentofacial Orthopedics, Dental School, University of Heidelberg, Im Neueheimer Feld 400, 69129 Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
45
|
Sakoda K, Yamamoto M, Negishi Y, Liao JK, Node K, Izumi Y. Simvastatin decreases IL-6 and IL-8 production in epithelial cells. J Dent Res 2006; 85:520-3. [PMID: 16723648 PMCID: PMC3001137 DOI: 10.1177/154405910608500608] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many cardiovascular studies have suggested that 3-hydroxy-3-methylglutaryl co-enzyme A reductase inhibitors (statins) have anti-inflammatory effects independent of cholesterol lowering. As a chronic inflammatory disease, periodontitis shares some mechanisms with atherosclerosis. Since oral epithelial cells participate importantly in periodontal inflammation, we measured simvastatin effects on interleukin-6 and interleukin-8 production by cultured human epithelial cell line (KB cells) in response to interleukin-1alpha. Simvastatin decreased production, an effect reversed by adding mevalonate or geranylgeranyl pyrophosphate, but not farnesyl pyrophosphate. Simvastatin was found to reduce NF-kappaB and AP-1 promoter activity in KB cells. Dominant-negative Rac1 severely inhibited interleukin-1alpha-induced NF-kappaB and AP-1 promoter activity. Our results may indicate an anti-inflammatory effect of simvastatin on human oral epithelial cells, apparently involving Rac1 GTPase inhibition.
Collapse
Affiliation(s)
- K Sakoda
- Department of Periodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Sakuragaoka 8-35-1, Kagoshima 890-8544, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
BACKGROUND Reactive oxygen species are produced in a highly localized and specific pattern in biological stress responses. The present review examines the redox regulatory aspects of a number of molecular stress response mechanisms in both prokaryotes and eukaryotes. SCOPE The present review provides examples representing both the cytoplasmic stress response, often studied as the heat shock response, as well as the stress response of the endoplasmic reticulum, known as the unfolded protein response. The examples have been selected to illustrate the variety of ways that redox signals mediate and affect stress responses. CONCLUSIONS Redox regulatory mechanisms are intricately embedded in both the cytoplasmic and endoplasmic reticulum stress responses at multiple levels. Many different stimuli, both internal and external, activate endogenous production of reactive oxygen species as a necessary part of the intracellular communication system that activates stress responses.
Collapse
Affiliation(s)
- Nina Fedoroff
- Huck Institutes of the Life Sciences and Biology Department, Penn State University, University Park, PA 16803, USA.
| |
Collapse
|
47
|
Ruse M, Knaus UG. New players in TLR-mediated innate immunity: PI3K and small Rho GTPases. Immunol Res 2006; 34:33-48. [PMID: 16720897 DOI: 10.1385/ir:34:1:33] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 12/12/2022]
Abstract
Toll-like receptors (TLRs) play a crucial role in the innate immune system as a first line of defense against pathogens. TLR activation in phagocytes produces pro-inflammatory cytokines and chemokines that contribute directly to elimination of infectious agents and activation of adaptive immune responses. However, a sustained inflammatory response can result in tissue damage and generalized sepsis. This review summarizes the complex and sometimes conflicting links of TLR signaling with two important regulators of immune cells functions: phosphoinositide 3-kinases (PI3Ks) and small GTPases of the Rho family. A unified model of hierarchical organization of these signaling participants is still premature, given that the tools for delineating how control of TLRmediated pathways is achieved are just emerging. Critical progress in our understanding of spatial-temporal propagation of TLR signaling will certainly be provided in the near future by pharmacological targeting of PI3Ks using recently characterized, second-generation PI3K inhibitors in combination with gene-targeting strategies for PI3K subunits and Rho GTPases targeted to the murine myeloid compartment.
Collapse
Affiliation(s)
- Monica Ruse
- Department of Immunology, The Scripps Research Institute La Jolla, California.
| | | |
Collapse
|
48
|
Li Q, Harraz MM, Zhou W, Zhang LN, Ding W, Zhang Y, Eggleston T, Yeaman C, Banfi B, Engelhardt JF. Nox2 and Rac1 regulate H2O2-dependent recruitment of TRAF6 to endosomal interleukin-1 receptor complexes. Mol Cell Biol 2006; 26:140-54. [PMID: 16354686 PMCID: PMC1317618 DOI: 10.1128/mcb.26.1.140-154.2006] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Reactive oxygen species (ROS) generated by NADPH oxidases (Nox) have been implicated in the regulation of signal transduction. However, the cellular mechanisms that link Nox activation with plasma membrane receptor signaling remain poorly defined. We have found that Nox2-derived ROS influence the formation of an active interleukin-1 (IL-1) receptor complex in the endosomal compartment by directing the H2O2-dependent binding of TRAF6 to the IL-1R1/MyD88 complex. Clearance of both superoxide and H2O2 from within the endosomal compartment significantly abrogated IL-1beta-dependent IKK and NF-kappaB activation. MyD88-dependent endocytosis of IL-1R1 following IL-1beta binding was required for the redox-dependent formation of an active endosomal receptor complex competent for IKK and NF-kappaB activation. Small interfering RNAs to either MyD88 or Rac1 inhibited IL-1beta induction of endosomal superoxide and NF-kappaB activation. However, MyD88 and Rac1 appear to be recruited independently to IL-1R1 following ligand stimulation. In this context, MyD88 binding was required for inducing endocytosis of IL-1R1 following ligand binding, while Rac1 facilitated the recruitment of Nox2 into the endosomal compartment and subsequent redox-dependent recruitment of TRAF6 to the MyD88/IL-1R1 complex. The identification of Nox-active endosomes helps explain how subcellular compartmentalization of redox signals can be used to direct receptor activation from the plasma membrane.
Collapse
Affiliation(s)
- Qiang Li
- Department of Anatomy and Cell Biology, College of Medicine, University of Iowa, 51 Newton Road, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Li Q, Engelhardt JF. Interleukin-1beta induction of NFkappaB is partially regulated by H2O2-mediated activation of NFkappaB-inducing kinase. J Biol Chem 2006; 281:1495-505. [PMID: 16286467 DOI: 10.1074/jbc.m511153200] [Citation(s) in RCA: 166] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Reactive oxygen species (ROS) have been demonstrated to act as second messengers in a number of signal transduction pathways, including NFkappaB. However, the mechanism(s) by which ROS regulate NFkappaB remain unclear and controversial. In the present report, we describe a mechanism whereby interleukin-1beta (IL-1beta) stimulation of NFkappaB is partially regulated by H2O2-mediated activation of NIK and subsequent NIK-mediated phosphorylation of IKKalpha. IL-1beta induced H2O2 production in MCF-7 cells and clearance of this ROS through the expression of GPx-1 reduced NFkappaB transcriptional activation by inhibiting NIK-mediated phosphorylation of IKKalpha. Although IKKalpha and IKKbeta were both involved in IL-1beta-mediated activation of NFkappaB, only the IKKalpha-dependent component was modulated by changes in H2O2 levels. Interestingly, in vitro reconstitution experiments demonstrated that NIK was activated by a very narrow range of H2O2 (1-10 microM), whereas higher concentrations (100 microM to 1 mM) inhibited NIK activity. Treatment of cells with the general Ser/Thr phosphatase inhibitor (okadaic acid) lead to activation of NFkappaB and enhanced NIK activity as a IKKalpha kinase, suggesting that ROS may directly regulate NIK through the inhibition of phosphatases. Recruitment of NIK to TRAF6 following IL-1beta stimulation was inhibited by H2O2 clearance and Rac1 siRNA, suggesting that Rac-dependent NADPH oxidase may be a source of ROS required for NIK activation. In summary, our studies have demonstrated that redox regulation of NIK by H2O2 is mechanistically important in IL-1beta induction of NFkappaB activation.
Collapse
Affiliation(s)
- Qiang Li
- Department of Anatomy & Cell Biology, College of Medicine, The University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
50
|
Schmeck B, Huber S, Moog K, Zahlten J, Hocke AC, Opitz B, Hammerschmidt S, Mitchell TJ, Kracht M, Rosseau S, Suttorp N, Hippenstiel S. Pneumococci induced TLR- and Rac1-dependent NF-kappaB-recruitment to the IL-8 promoter in lung epithelial cells. Am J Physiol Lung Cell Mol Physiol 2005; 290:L730-L737. [PMID: 16299055 DOI: 10.1152/ajplung.00271.2005] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Streptococcus pneumoniae is the major pathogen of community-acquired pneumonia. The respiratory epithelium constitutes the first line of defense against invading lung pathogens, including pneumococci. We analyzed the involvement of Toll-like receptors (TLR) and Rho-GTPase signaling in the activation of human lung epithelial cells by pneumococci. S. pneumoniae induced release of interleukin-8 (IL-8) by human bronchial epithelial cell line BEAS-2B. Specific inhibition of Rac1 by Nsc23766 or a dominant-negative mutant of Rac1 strongly reduced cytokine release. In addition, pneumococci-related cell activation (IL-8 release, NF-kappaB-activation) depended on MyD88, phosphatidylinositol 3-kinase, and Cdc42 but not on RhoA. Pneumococci enhanced TLR1 and TLR2 mRNA expression in BEAS-2B cells, whereas TLR4 and TLR6 expression was constitutively high. TLR1 and 2 synergistically recognized pneumococci in cotransfection experiments. TLR4, TLR6, LPS-binding protein, and CD14 seem not to be involved in pneumococci-dependent cell activation. At the IL-8 gene promoter, recruitment of phosphorylated NF-kappaB subunit p65 was blocked by inhibition of Rac1, whereas binding of the phosphorylated activator protein-1 subunit c-Jun to the promoter was not diminished. In summary, these results suggest that S. pneumoniae activate human epithelial cells by TLR1/2 and a phosphatidylinositol 3-kinase- and Rac1-dependent NF-kappaB-recruitment to the IL-8 promoter.
Collapse
Affiliation(s)
- Bernd Schmeck
- Department of Internal Medicine/Infectious Diseases, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|