scholarly journals Nrf2 Regulates Anti-Inflammatory A20 Deubiquitinase Induction by LPS in Macrophages in Contextual Manner

Antioxidants ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 847
Author(s):  
Haranatha R. Potteti ◽  
Lalith K. Venkareddy ◽  
Patrick M. Noone ◽  
Aparna Ankireddy ◽  
Chandramohan R. Tamatam ◽  
...  

The aberrant regulation of inflammatory gene transcription following oxidant and inflammatory stimuli can culminate in unchecked systemic inflammation leading to organ dysfunction. The Nrf2 transcription factor dampens cellular stress and controls inflammation by upregulating antioxidant gene expression and TNFα-induced Protein 3 (TNFAIP3, aka A20) deubiquitinase by controlling NF-kB signaling dampens tissue inflammation. Here, we report that Nrf2 is required for A20 induction by inflammatory stimuli LPS in monocyte/bone marrow derived macrophages (MDMΦs) but not in lung-macrophages (LDMΦs). LPS-induced A20 expression was significantly lower in Nrf2−/− MDMΦs and was not restored by antioxidant supplementation. Nrf2 deficiency markedly impaired LPS-stimulated A20 mRNA expression Nrf2−/− MDMΦs and ChIP assays showed Nrf2 enrichment at the promoter Nrf2−/− MDMΦs upon LPS stimulation, demonstrating that Nrf2 directly regulates A20 expression. Contrary to MDMΦs, LPS-stimulated A20 expression was not largely impaired in Nrf2−/− LDMΦs ex vivo and in vivo and ChIP assays showed lack of increased Nrf2 binding at the A20 promoter in LDMΦ following LPS treatment. Collectively, these results demonstrate a crucial role for Nrf2 in optimal A20 transcriptional induction in macrophages by endotoxin, and this regulation occurs in a contextual manner.

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Allen Volchuk ◽  
Anna Ye ◽  
Leon Chi ◽  
Benjamin E. Steinberg ◽  
Neil M. Goldenberg

Abstract The protein high-mobility group box 1 (HMGB1) is released into the extracellular space in response to many inflammatory stimuli, where it is a potent signaling molecule. Although research has focused on downstream HMGB1 signaling, the means by which HMGB1 exits the cell is controversial. Here we demonstrate that HMGB1 is not released from bone marrow-derived macrophages (BMDM) after lipopolysaccharide (LPS) treatment. We also explore whether HMGB1 is released via the pore-forming protein gasdermin D after inflammasome activation, as is the case for IL-1β. HMGB1 is only released under conditions that cause cell lysis (pyroptosis). When pyroptosis is prevented, HMGB1 is not released, despite inflammasome activation and IL-1β secretion. During endotoxemia, gasdermin D knockout mice secrete HMGB1 normally, yet secretion of IL-1β is completely blocked. Together, these data demonstrate that in vitro HMGB1 release after inflammasome activation occurs after cellular rupture, which is probably inflammasome-independent in vivo.


1993 ◽  
Vol 75 (3) ◽  
pp. 1256-1262 ◽  
Author(s):  
S. C. Erzurum ◽  
C. Danel ◽  
A. Gillissen ◽  
C. S. Chu ◽  
B. C. Trapnell ◽  
...  

Human bronchial epithelium is exquisitely sensitive to high O2 levels, with tracheobronchitis usually developing after 12 h of exposure to 100% O2. To evaluate whether this vulnerability results from inability of the bronchial epithelium to provide adequate antioxidant protection, we quantified antioxidant gene expression in bronchial epithelium of normal volunteers at baseline and after exposure to 100% O2 in vivo. After 14.8 +/- 0.2 h of 100% O2, 24 of 33 individuals had evidence of tracheobronchitis. Baseline gene expression of CuZn superoxide dismutase (SOD), MnSOD, and catalase in bronchial epithelium was very low (CuZnSOD 4.1 +/- 0.8 transcripts/cell, MnSOD 5.1 +/- 0.9, catalase 1.3 +/- 0.2), with control gamma-actin expression relatively abundant (50 +/- 6 transcripts/cell). Importantly, despite 100% O2 exposure sufficient to cause tracheobronchitis in most individuals, antioxidant mRNA transcripts/cell in bronchial epithelium did not increase (P > 0.5). Catalase activity in bronchial epithelium did not change after exposure to hyperoxia (P > 0.05). Total SOD activity increased mildly (P < 0.01) but not sufficiently to protect the epithelium. Together, the very low levels of expression of intracellular antioxidant enzymes and the inability to upregulate expression at the mRNA level with oxidant stress likely have a role in human airway epithelium susceptibility to hyperoxia.


2008 ◽  
Vol 103 (1) ◽  
Author(s):  
Henna-Kaisa Jyrkkänen ◽  
Emilia Kansanen ◽  
Matias Inkala ◽  
Annukka M. Kivelä ◽  
Hanna Hurttila ◽  
...  

2020 ◽  
Vol 11 (1) ◽  
Author(s):  
Hao Daniel Lin ◽  
Chui-Yee Fong ◽  
Arijit Biswas ◽  
Ariff Bongso

Abstract Background The transplantation of human umbilical cord blood (UCB) CD34+ cells has been successfully used to treat hematological disorders but one major limitation has been the low cell numbers available. Mesenchymal stem cells (MSCs) lying within the bone marrow in vivo behave like a scaffold on which CD34+ cells interact and proliferate. We therefore evaluated the use of allogeneic MSCs from the human UC Wharton’s jelly (hWJSCs) as stromal support for the ex vivo expansion of CD34+ cells. Methods We performed an in-depth evaluation of the primitiveness, migration, adhesion, maturation, mitochondrial behavior, and pathway mechanisms of this platform using conventional assays followed by the evaluation of engraftment potential of the expanded CD34+ cells in an in vivo murine model. Results We demonstrate that hWJSCs and its conditioned medium (hWJSC-CM) support the production of significantly high fold changes of CD34+, CD34+CD133+, CD34+CD90+, CD34+ALDH+, CD34+CD45+, and CD34+CD49f+ cells after 7 days of interaction when compared to controls. In the presence of hWJSCs or hWJSC-CM, the CD34+ cells produced significantly more primitive CFU-GEMM colonies, HoxB4, and HoxA9 gene expression and lower percentages of CD34+CXCR4+ cells. There were also significantly higher N-cadherin+ cell numbers and increased cell migration in transwell migration assays. The CD34+ cells expanded with hWJSCs had significantly lower mitochondrial mass, mitochondrial membrane potential, and oxidative stress. Green Mitotracker-tagged mitochondria from CD34+ cells were observed lying within red CellTracker-tagged hWJSCs under confocal microscopy indicating mitochondrial transfer via tunneling nanotubes. CD34+ cells expanded with hWJSCs and hWJSC-CM showed significantly reduced oxidative phosphorylation (ATP6VIH and NDUFA10) and increased glycolytic (HIF-1a and HK-1) pathway-related gene expression. CD34+ cells expanded with hWJSCs for 7 days showed significant greater CD45+ cell chimerism in the bone marrow of primary and secondary irradiated mice when transplanted intravenously. Conclusions In this report, we confirmed that allogeneic hWJSCs provide an attractive platform for the ex vivo expansion of high fold numbers of UCB CD34+ cells while keeping them primitive. Allogeneic hWJSCs are readily available in abundance from discarded UCs, can be easily frozen in cord blood banks, thawed, and then used as a platform for UCB-HSC expansion if numbers are inadequate.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1715-1715
Author(s):  
Craig E. Eckfeldt ◽  
Eric M. Mendenhall ◽  
Catherine M. Verfaillie

Abstract Hematopoietic stem cells (HSCs) are functionally defined by their capacity to home to the bone marrow microenvironment, proliferate and differentiate to restore normal hematopoiesis in a myeloablated recipient; however, the molecular determinants of these processes are not well understood. By comparing the gene expression profiles of highly purified human HSC-enriched and HSC-depleted cell populations, and subsequently validating the hematopoietic function of a subset of these differentially expressed genes using zebrafish (Danio rerio), we previously identified human Sprouty 1 (SPRY1), an evolutionarily conserved antagonist of RTK signaling, as a potential regulator of mammalian HSC development and/or cell-fate decisions. To directly assess the role of SPRY1 in mammalian HSC and hematopoietic progenitor cell (HPC) function, we constructed a dual-promoter lentiviral vector to co-express SPRY1 and green fluorescent protein (GFP) (SPRY1-LV) and a control lentiviral vector to express GFP alone (GFP-LV) in the CD34+ fraction of human umbilical cord blood (UCB). While the enforced expression of SPRY1 in CD34+ UCB cells had no effect on the frequency or morphology of colonies generated in short-term in vitro colony-forming cell (CFC) assays (SPRY1-LV = 27.6 ± 12.3% and GFP-LV = 28.4 ± 20.0%; n = 3), it profoundly inhibited the capacity of UCB CD34+ cells to engraft in the bone marrow NOD-SCID mice in vivo. The estimated frequency of week 11 SCID-repopulating cells (SRC) (± 1 standard error) for SPRY1-LV (n = 13 mice) and GFP-LV (n = 15 mice) cells was 1 in 12,678 cells (6,167 – 26,064) and 1 in 3,412 cells (2,272 – 5,124), respectively, as determined using limiting dilution conditions and Poisson statistics. Furthermore, in 14 day cultures designed for the ex vivo expansion and/or maintenance of primitive hematopoietic cells, ectopic expression of SPRY1 in CD34+ UCB cells dramatically inhibited the expansion of total nucleated cells (SPRY1-LV = 37.9 ± 10.1 fold; GFP-LV = 71.1 ± 5.8 fold; n = 3; p&lt;0.05) and CFCs (SPRY1-LV = 5.7 ± 1.2 fold; GFP-LV = 20.8 ± 14.9 fold; n = 3), although it had no effect on expansion of CD34+CD38− cells. We are currently investigating potential mechanisms for the observed affects of SPRY1 on primitive hematopoietic cells, paying particular attention to the possible effects of SPRY1 expression on “early-acting” hematopoietic cytokines and growth factors that activate RTKs - including FGF1, FGF2, VEGF, SCF, FLT3L, and ANGPT1. In conclusion, enforced expression of SPRY1 negatively regulates primitive hematopoietic cell engraftment in vivo and expansion in vitro, thereby presenting the first example of a role for a Sprouty family member, SPRY1, in primitive human hematopoietic cell function. Moreover, this data further validates the use of model organisms, such as zebrafish, for evaluating the functional roles of transcripts identified in large-scale gene expression profiling experiments in mice and humans.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1034-1034
Author(s):  
Robert A. Campbell ◽  
Jesse W Rowley ◽  
Andrew S. Weyrich ◽  
Matthew T. Rondina

Abstract Background IFITM proteins (IFITM-1, -2, and -3) mediate cellular resistance to influenza, dengue, and other viruses. IFITM expression on human platelets has not been previously recognized. Our laboratory recently demonstrated that IFITMs are robustly expressed by human platelets and megakaryocytes after stimulation by pathogens and inflammatory mediators and restrict viral infection. IFITMs, which are interferon inducible, also mediate clathrin localization and associated protein endocytosis. Nevertheless, whether IFITMs regulate protein endocytosis by platelets and megakaryocytes remains unknown. Aims We investigated IFITM expression on murine megakaryocytes and platelets and determined whether IFITMs regulate fibrinogen endocytosis under basal and inflammatory conditions. Methods We examined the expression of IFITMs and clathrin in bone-marrow derived murine megakaryocytes and platelets under basal conditions and following interferon-beta (IFN-β) stimulation. To determine whether upregulation of IFITM causes increased fibrinogen endocytosis, megakaryocytes were stimulated ex vivo with IFN-β and treated with labeled fibrinogen. Endocytosis of labeled fibrinogen was then measured by immunocytochemistry and flow cytometry. To determine whether this response also occurred in vivo, C57Bl/6 mice were injected intraperitoneally (IP) with 50,000 units of IFN-β over four days. On the fourth day, 100 μg of labeled fibrinogen was injected into the tail vein and the amount of endocytosed, labeled fibrinogen in platelets was determined the next day via flow cytometry. Parallel experiments were performed in age and gender matched IFITM-/- mice. Results Bone-marrow derived murine megakaryocytes and platelets basally express IFITMs. Upon IFN-β stimulation, IFITM and clathrin expression significantly increased (p<0.05). Fibrinogen endocytosis by murine megakaryocytes occurred under resting conditions and appeared to be punctate and granular in nature. Upon IFN-b stimulation, fibrinogen endocytosis in megakaryocytes significantly increased compared to unstimulated conditions (p<0.004). The increase in endocytosis appeared independent of changes in αIIbβ3 expression as IFN-β stimulation did not change αIIbβ3 surface protein. Fibrinogen endocytosis after IFN-β stimulation did not increase in megakaryocytes from IFITM-/- mice, suggesting that IFITMs regulate fibrinogen uptake under these conditions. We next determined if fibrinogen endocytosis occurred in platelets isolated from IFITM-/- mice. Platelet counts and activation indices (assessed by JonA staining) were similar in C57Bl/6 mice (WT) and IFITM-/- mice. Nevertheless, the injection of IFN-β IP results in significant increases in fibrinogen endocytosis by platelets in vivo in WT but not IFITM-/- mice (p<0.02). Summary/Conclusions These findings suggest IFITMs, in addition to their anti-viral roles, mediate fibrinogen endocytosis. Further, in settings where inflammatory stimuli such as interferons are increased, enhanced IFITM expression may promote upregulation of fibrinogen endocytosis by platelets and megakaryocytes. Disclosures No relevant conflicts of interest to declare.


2009 ◽  
Vol 106 (6) ◽  
pp. 1935-1942 ◽  
Author(s):  
Farnaz P. Baqai ◽  
Daila S. Gridley ◽  
James M. Slater ◽  
Xian Luo-Owen ◽  
Louis S. Stodieck ◽  
...  

Spaceflight conditions have a significant impact on a number of physiological functions due to psychological stress, radiation, and reduced gravity. To explore the effect of the flight environment on immunity, C57BL/6NTac mice were flown on a 13-day space shuttle mission (STS-118). In response to flight, animals had a reduction in liver, spleen, and thymus masses compared with ground (GRD) controls ( P < 0.005). Splenic lymphocyte, monocyte/macrophage, and granulocyte counts were significantly reduced in the flight (FLT) mice ( P < 0.05). Although spontaneous blastogenesis of splenocytes in FLT mice was increased, response to lipopolysaccharide (LPS), a B-cell mitogen derived from Escherichia coli, was decreased compared with GRD mice ( P < 0.05). Secretion of IL-6 and IL-10, but not TNF-α, by LPS-stimulated splenocytes was increased in FLT mice ( P < 0.05). Finally, many of the genes responsible for scavenging reactive oxygen species were upregulated after flight. These data indicate that exposure to the spaceflight environment can increase anti-inflammatory mechanisms and change the ex vivo response to LPS, a bacterial product associated with septic shock and a prominent Th1 response.


Sign in / Sign up

Export Citation Format

Share Document