Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Sensors (Basel) ; 22(9)2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35590971

ABSTRACT

Compared to other types of sensors, fiber optic sensors have improved accuracy and durability. Recently, the Smart Strand was developed to maximize the advantages of fiber optic sensors for measuring the cable forces in prestressed concrete structures or cable-supported bridges. The Smart Strand has fiber Bragg gratings (FBGs) embedded in a core wire of the seven-wire strand. Similar to other sensors, the strain measured at an FBG is affected by temperature; therefore, the temperature effect that is not related to the mechanical strain should be compensated for or corrected in the long-term measurement subjected to temperature variation. However, a temperature compensation procedure for the FBG has yet to be established, and relevant studies have used different formulas for the compensation. Moreover, when the FBG sensors are packaged with a certain material-such as fiber reinforced polymer-for protection, it is important to consider the interaction between the FBG, packaging material, and host material during thermal behavior. Therefore, this study proposed a reasonable procedure for temperature compensation for the FBG sensors embedded in packaging material and host material. In particular, the thermal sensitivity of the Smart Strand was intensively investigated. The proposed theoretical formulas were validated through comparison with data obtained from various specimens in a temperature-controlled chamber. Finally, the procedure was applied to correct the data measured using the Smart Strands in a 20-m-long full-scale specimen for about a year, thus resulting in a realistic trend of the long-term prestressing force.


Subject(s)
Fiber Optic Technology , Optical Fibers , Fiber Optic Technology/methods , Mechanical Phenomena , Temperature
2.
Circulation ; 142(18): 1736-1751, 2020 11 03.
Article in English | MEDLINE | ID: mdl-32883094

ABSTRACT

BACKGROUND: Macrophages produce many inflammation-associated molecules, released by matrix metalloproteinases, such as adhesion molecules, and cytokines, as well, which play a crucial role in atherosclerosis. In this context, we investigated the relationship between Ninjurin-1 (Ninj1 [nerve injury-induced protein]), a novel matrix metalloproteinase 9 substrate, expression, and atherosclerosis progression. METHODS: Ninj1 expression and atherosclerosis progression were assessed in atherosclerotic aortic tissue and serum samples from patients with coronary artery disease and healthy controls, and atheroprone apolipoprotein e-deficient (Apoe-/-) and wild-type mice, as well. Apoe-/- mice lacking systemic Ninj1 expression (Ninj1-/-Apoe-/-) were generated to assess the functional effects of Ninj1. Bone marrow transplantation was also used to generate low-density lipoprotein receptor-deficient (Ldlr-/-) mice that lack Ninj1 specifically in bone marrow-derived cells. Mice were fed a Western diet for 5 to 23 weeks, and atherosclerotic lesions were investigated. The anti-inflammatory role of Ninj1 was verified by treating macrophages and mice with the peptides Ninj11-56 (ML56) and Ninj126-37 (PN12), which mimic the soluble form of Ninj1 (sNinj1). RESULTS: Our in vivo results conclusively showed a correlation between Ninj1 expression in aortic macrophages and the extent of human and mouse atherosclerotic lesions. Ninj1-deficient macrophages promoted proinflammatory gene expression by activating mitogen-activated protein kinase and inhibiting the phosphoinositide 3-kinase/Akt signaling pathway. Whole-body and bone marrow-specific Ninj1 deficiencies significantly increased monocyte recruitment and macrophage accumulation in atherosclerotic lesions through elevated macrophage-mediated inflammation. Macrophage Ninj1 was directly cleaved by matrix metalloproteinase 9 to generate a soluble form that exhibited antiatherosclerotic effects, as assessed in vitro and in vivo. Treatment with the sNinj1-mimetic peptides, ML56 and PN12, reduced proinflammatory gene expression in human and mouse classically activated macrophages, thereby attenuating monocyte transendothelial migration. Moreover, continuous administration of mPN12 alleviated atherosclerosis by inhibiting the enhanced monocyte recruitment and inflammation characteristics of this disorder in mice, regardless of the presence of Ninj1. CONCLUSIONS: Ninj1 is a novel matrix metalloproteinase 9 substrate in macrophages, and sNinj1 is a secreted atheroprotective protein that regulates macrophage inflammation and monocyte recruitment in atherosclerosis. Moreover, sNinj1-mediated anti-inflammatory effects are conserved in human macrophages and likely contribute to human atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atherosclerosis , Cell Adhesion Molecules, Neuronal , Macrophages/metabolism , Nerve Growth Factors , Peptidomimetics/pharmacology , Signal Transduction/drug effects , Animals , Atherosclerosis/drug therapy , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion Molecules, Neuronal/pharmacology , Female , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout, ApoE , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics
3.
FASEB J ; 34(6): 8702-8720, 2020 06.
Article in English | MEDLINE | ID: mdl-32385864

ABSTRACT

Disruption of colonic homeostasis caused by aberrant M1/M2 macrophage polarization and dysbiosis contributes to inflammatory bowel disease (IBD) pathogenesis. However, the molecular factors mediating colonic homeostasis are not well characterized. Here, we found that Ninjurin1 (Ninj1) limits colon inflammation by regulating macrophage polarization and microbiota composition under homeostatic conditions and during colitis development. Ninj1 deletion in mice induced hypersusceptibility to colitis, with increased prevalence of colitogenic Prevotellaceae strains and decreased immunoregulatory Lachnospiraceae strains. Upon co-housing (CoH) with WT mice, Ninj1-/- mice showed increased Lachnospiraceae and decreased Prevotellaceae abundance, with subsequent improvement of colitis. Under homeostatic conditions, M1 macrophage frequency was higher in the Ninj1-/- mouse colons than wild-type (WT) mouse colons, which may contribute to increased basal colonic inflammation and microbial imbalance. Following colitis induction, Ninj1 expression was increased in macrophages; meanwhile Ninj1-/- mice showed severe colitis development and impaired recovery, associated with decreased M2 macrophages and escalated microbial imbalance. In vitro, Ninj1 knockdown in mouse and human macrophages activated M1 polarization and restricted M2 polarization. Finally, the transfer of WT macrophages ameliorated severe colitis in Ninj1-/- mice. These findings suggest that Ninj1 mediates colonic homeostasis by modulating M1/M2 macrophage balance and preventing extensive dysbiosis, with implications for IBD prevention and therapy.


Subject(s)
Cell Adhesion Molecules, Neuronal/deficiency , Colitis/metabolism , Colitis/pathology , Gastrointestinal Microbiome/physiology , Macrophages/metabolism , Macrophages/pathology , Nerve Growth Factors/deficiency , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Colon/pathology , Disease Models, Animal , Homeostasis/physiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Macrophage Activation/physiology , Male , Mice , Nerve Growth Factors/metabolism , THP-1 Cells/metabolism
4.
Proc Natl Acad Sci U S A ; 114(10): E1885-E1894, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223506

ABSTRACT

CpG, 5'-C-phosphate-G-3', islands (CGIs) have long been known for their association with enhancers, silencers, and promoters, and for their epigenetic signatures. They are maintained in embryonic stem cells (ESCs) in a poised but inactive state via the formation of bivalent chromatin containing both active and repressive marks. CGIs also occur within coding sequences, where their functional role has remained obscure. Intragenic CGIs (iCGIs) are largely absent from housekeeping genes, but they are found in all genes associated with organ development and cell lineage control. In this paper, we investigated the epigenetic status of iCGIs and found that they too reside in bivalent chromatin in ESCs. Cell type-specific DNA methylation of iCGIs in differentiated cells was linked to the loss of both the H3K4me3 and H3K27me3 marks, and disruption of physical interaction with promoter regions, resulting in transcriptional activation of key regulators of differentiation such as PAXs, HOXs, and WNTs. The differential epigenetic modification of iCGIs appears to be mediated by cell type-specific transcription factors distinct from those bound by promoter, and these transcription factors may be involved in the hypermethylation of iCGIs upon cell differentiation. iCGIs thus play a key role in the cell type-specific regulation of transcription.


Subject(s)
Cell Differentiation/genetics , CpG Islands/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Cell Lineage/genetics , Chromatin/genetics , Embryonic Stem Cells/cytology , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Histones/genetics , Humans , Promoter Regions, Genetic
6.
J Biol Chem ; 289(32): 21926-36, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24917672

ABSTRACT

Ninjurin1 is involved in the pathogenesis of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis, by mediating leukocyte extravasation, a process that depends on homotypic binding. However, the precise regulatory mechanisms of Ninjurin1 during inflammation are largely undefined. We therefore examined the pro-migratory function of Ninjurin1 and its regulatory mechanisms in macrophages. Interestingly, Ninjurin1-deficient bone marrow-derived macrophages exhibited reduced membrane protrusion formation and dynamics, resulting in the impairment of cell motility. Furthermore, exogenous Ninjurin1 was distributed at the membrane of filopodial structures in Raw264.7 macrophage cells. In Raw264.7 cells, RNA interference of Ninjurin1 reduced the number of filopodial projections, whereas overexpression of Ninjurin1 facilitated their formation and thus promoted cell motility. Ninjurin1-induced filopodial protrusion formation required the activation of Rac1. In Raw264.7 cells penetrating an MBEC4 endothelial cell monolayer, Ninjurin1 was localized to the membrane of protrusions and promoted their formation, suggesting that Ninjurin1-induced protrusive activity contributed to transendothelial migration. Taking these data together, we conclude that Ninjurin1 enhances macrophage motility and consequent extravasation of immune cells through the regulation of protrusive membrane dynamics. We expect these findings to provide insight into the understanding of immune responses mediated by Ninjurin1.


Subject(s)
Cell Adhesion Molecules, Neuronal/physiology , Cell Movement/physiology , Macrophages/physiology , Nerve Growth Factors/physiology , Animals , Cell Adhesion/physiology , Cell Adhesion Molecules, Neuronal/deficiency , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Cell Membrane/physiology , Cells, Cultured , Endothelial Cells/physiology , Gene Knockdown Techniques , Inflammation/etiology , Inflammation/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nerve Growth Factors/deficiency , Nerve Growth Factors/genetics , Neuropeptides/metabolism , Pseudopodia/physiology , RNA Interference , rac1 GTP-Binding Protein/metabolism
7.
J Biol Chem ; 289(6): 3328-38, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24347169

ABSTRACT

Ninjurin1 is a homotypic adhesion molecule that contributes to leukocyte trafficking in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, in vivo gene deficiency animal studies have not yet been done. Here, we constructed Ninjurin1 knock-out (KO) mice and investigated the role of Ninjurin1 on leukocyte trafficking under inflammation conditions such as EAE and endotoxin-induced uveitis. Ninjurin1 KO mice attenuated EAE susceptibility by reducing leukocyte recruitment into the injury regions of the spinal cord and showed less adhesion of leukocytes on inflamed retinal vessels in endotoxin-induced uveitis mice. Moreover, the administration of a custom-made antibody (Ab26-37) targeting the Ninjurin1 binding domain ameliorated the EAE symptoms, showing the contribution of its adhesion activity to leukocyte trafficking. In addition, we addressed the transendothelial migration (TEM) activity of bone marrow-derived macrophages and Raw264.7 cells according to the expression level of Ninjurin1. TEM activity was decreased in Ninjurin1 KO bone marrow-derived macrophages and siNinj1 Raw264.7 cells. Consistent with this, GFP-tagged mNinj1-overexpressing Raw264.7 cells increased their TEM activity. Taken together, we have clarified the contribution of Ninjurin1 to leukocyte trafficking in vivo and delineated its direct functions to TEM, emphasizing Ninjurin1 as a beneficial therapeutic target against inflammatory diseases such as multiple sclerosis.


Subject(s)
Bone Marrow Cells/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement , Encephalomyelitis, Autoimmune, Experimental/metabolism , Macrophages/metabolism , Nerve Growth Factors/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Bone Marrow Cells/pathology , Cell Adhesion Molecules, Neuronal/antagonists & inhibitors , Cell Adhesion Molecules, Neuronal/genetics , Cell Line , Disease Susceptibility , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/therapy , Macrophages/pathology , Mice , Mice, Knockout , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Multiple Sclerosis/therapy , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/genetics
8.
FASEB J ; 28(11): 4779-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25059229

ABSTRACT

CD137 (4-1BB), a member of the tumor necrosis factor receptor superfamily, has been reported to be expressed in atherosclerotic plaques, and to promote lesion formation. However, the role of CD137 in mediating atherosclerotic plaque stability and the possible underlying molecular and cellular mechanisms are poorly understood. Here, apolipoprotein E-deficient (ApoE(-/-)) and CD137-deficient ApoE(-/-) (ApoE(-/-)CD137(-/-)) mice fed a chow diet for 66 wk were used. CD137 induces plaque instability, which is characterized by increased plaque necrosis, decreased collagen content, decreased vascular smooth muscle cell (VSMC) content, and increased macrophage infiltration. CD137 also increases the infiltration of effector T (Teff) cells into plaque lesion sites, resulting in increased interferon-γ (IFN-γ) expression. Interestingly, Teff-cell-derived IFN-γ inhibits collagen synthesis in atherosclerotic plaques. Furthermore, CD137 activation increases the apoptosis of VSMCs, possibly by decreasing the antiapoptotic regulator, Bcl-2, and subsequently up-regulating cleaved caspase-3. In macrophages, activation of CD137 signaling boosted the oxidized low density lipoprotein-induced expression of matrix metalloproteinase 9 via the p38 mitogen-activated protein kinase and extracellular signal-regulated kinase1/2 signaling pathways. In summary, activation of CD137 signaling decreases the stability of advanced atherosclerotic plaques via its combined effects on Teff cells, VSMCs, and macrophages.


Subject(s)
4-1BB Ligand/immunology , Atherosclerosis/metabolism , Hyperlipidemias/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , T-Lymphocytes/metabolism , Animals , Apoptosis/drug effects , Atherosclerosis/immunology , Interferon-gamma/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology
9.
Mediators Inflamm ; 2015: 492659, 2015.
Article in English | MEDLINE | ID: mdl-26576074

ABSTRACT

Initial and recurrent stroke produces central nervous system (CNS) damage, involving neuroinflammation. Receptor-mediated S1P signaling can influence neuroinflammation and has been implicated in cerebral ischemia through effects on the immune system. However, S1P-mediated events also occur within the brain itself where its roles during stroke have been less well studied. Here we investigated the involvement of S1P signaling in initial and recurrent stroke by using a transient middle cerebral artery occlusion/reperfusion (M/R) model combined with analyses of S1P signaling. Gene expression for S1P receptors and involved enzymes was altered during M/R, supporting changes in S1P signaling. Direct S1P microinjection into the normal CNS induced neuroglial activation, implicating S1P-initiated neuroinflammatory responses that resembled CNS changes seen during initial M/R challenge. Moreover, S1P microinjection combined with M/R potentiated brain damage, approximating a model for recurrent stroke dependent on S1P and suggesting that reduction in S1P signaling could ameliorate stroke damage. Delivery of FTY720 that removes S1P signaling with chronic exposure reduced damage in both initial and S1P-potentiated M/R-challenged brain, while reducing stroke markers like TNF-α. These results implicate direct S1P CNS signaling in the etiology of initial and recurrent stroke that can be therapeutically accessed by S1P modulators acting within the brain.


Subject(s)
Brain Ischemia/prevention & control , Lysophospholipids/physiology , Receptors, Lysosphingolipid/antagonists & inhibitors , Sphingosine/analogs & derivatives , Stroke/prevention & control , Animals , Blood-Brain Barrier , Brain Ischemia/etiology , Fingolimod Hydrochloride/pharmacology , Lysophospholipids/toxicity , Male , Mice , Mice, Inbred ICR , Microinjections , Neuroglia/drug effects , Neuroglia/physiology , Phosphotransferases (Alcohol Group Acceptor)/physiology , Signal Transduction , Sphingosine/physiology , Sphingosine/toxicity , Stroke/etiology , Tumor Necrosis Factor-alpha/analysis
10.
Redox Biol ; 54: 102347, 2022 08.
Article in English | MEDLINE | ID: mdl-35688114

ABSTRACT

Ischemic stroke is the leading cause of immortal disability and death worldwide. For treatment in the acute phase, it is necessary to control excessive reactive oxygen species (ROS) damage during ischemia/reperfusion (I/R). Microglia are well known to be closely associated with excessive ROS response in the early stage of I/R. However, the precise roles of microglia associated with mitigating ROS damage, and molecular markers of heterogenetic microglia in the I/R damaged brain has not been clarified. Here, we identified a new type of microglia associated with stroke in the I/R injured brain. Single-cell RNA sequencing (scRNA-seq) was used to assess transcriptional changes of microglia and immune cells in the contralateral (CL) and ipsilateral (IL) hemispheres after transient middle cerebral artery occlusion (tMCAO) surgery to mimic ischemic stroke. We classified a unique type of microglia with enhanced antioxidant function and markers similar to those of disease-associated microglia (DAM), designated them as stroke-associated microglia (SAM). The representative antioxidant enzyme, Peroxiredoxin-1 (Prdx1), was predominantly expressed in SAM and mediated ROS defense genes, including Txn1, Srx1, Mt1, and Mt2. In the Prdx1-/- I/R damaged brain, we observed significantly increased infarction, as assessed by TTC staining, and FACS analysis detected severe microglial cell death. Importantly, scRNA transcriptomics data showed that the SAM population was specifically decreased in Prdx1-/- mice and that these mice exhibited decreased ROS damage resistance. Inflammatory responses which were detected by ELISA and qPCR, were also increased in Prdx1-/- IL hemispheres. Finally, Prdx1-dependent antioxidative SAM were found to be essential for increasing the transcription levels of stroke-protective molecules, such as osteopontin and ferritin. A novel microglia type (SAM) is specifically activated in response to stroke I/R injury, and that Prdx1 expression is required for the activation and enhanced antioxidant function of SAM.


Subject(s)
Brain Ischemia , Ischemic Stroke , Peroxiredoxins , Stroke , Animals , Antioxidants/metabolism , Brain Ischemia/genetics , Brain Ischemia/metabolism , Ischemic Stroke/genetics , Mice , Microglia/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Stroke/genetics , Stroke/metabolism
11.
Nat Commun ; 13(1): 6647, 2022 11 04.
Article in English | MEDLINE | ID: mdl-36333342

ABSTRACT

Endothelial nitric oxide synthase (eNOS) decreases following inflammatory stimulation. As a master regulator of endothelial homeostasis, maintaining optimal eNOS levels is important during cardiovascular events. However, little is known regarding the mechanism of eNOS protection. In this study, we demonstrate a regulatory role for endothelial expression of 2'-5' oligoadenylate synthetase-like 1 (OASL1) in maintaining eNOS mRNA stability during athero-prone conditions and consider its clinical implications. A lack of endothelial Oasl1 accelerated plaque progression, which was preceded by endothelial dysfunction, elevated vascular inflammation, and decreased NO bioavailability following impaired eNOS expression. Mechanistically, knockdown of PI3K/Akt signaling-dependent OASL expression increased Erk1/2 and NF-κB activation and decreased NOS3 (gene name for eNOS) mRNA expression through upregulation of the negative regulatory, miR-584, whereas a miR-584 inhibitor rescued the effects of OASL knockdown. These results suggest that OASL1/OASL regulates endothelial biology by protecting NOS3 mRNA and targeting miR-584 represents a rational therapeutic strategy for eNOS maintenance in vascular disease.


Subject(s)
Atherosclerosis , MicroRNAs , Humans , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Ligases/metabolism , Endothelial Cells/metabolism , MicroRNAs/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , RNA, Messenger/metabolism , RNA Stability , Nitric Oxide/metabolism , Cells, Cultured
12.
Redox Biol ; 51: 102275, 2022 05.
Article in English | MEDLINE | ID: mdl-35248828

ABSTRACT

Mitochondrial quality control (MQC) consists of multiple processes: the prevention of mitochondrial oxidative damage, the elimination of damaged mitochondria via mitophagy and mitochondrial fusion and fission. Several studies proved that MQC impairment causes a plethora of pathological conditions including cardiovascular diseases. However, the precise molecular mechanism by which MQC reverses mitochondrial dysfunction, especially in the heart, is unclear. The mitochondria-specific peroxidase Peroxiredoxin 3 (Prdx3) plays a protective role against mitochondrial dysfunction by removing mitochondrial reactive oxygen species. Therefore, we investigated whether Prdx3-deficiency directly leads to heart failure via mitochondrial dysfunction. Fifty-two-week-old Prdx3-deficient mice exhibited cardiac hypertrophy and dysfunction with giant and damaged mitochondria. Mitophagy was markedly suppressed in the hearts of Prdx3-deficient mice compared to the findings in wild-type and Pink1-deficient mice despite the increased mitochondrial damage induced by Prdx3 deficiency. Under conditions inducing mitophagy, we identified that the damaged mitochondrial accumulation of PINK1 was completely inhibited by the ablation of Prdx3. We propose that Prdx3 interacts with the N-terminus of PINK1, thereby protecting PINK1 from proteolytic cleavage in damaged mitochondria undergoing mitophagy. Our results provide evidence of a direct association between MQC dysfunction and cardiac function. The dual function of Prdx3 in mitophagy regulation and mitochondrial oxidative stress elimination further clarifies the mechanism of MQC in vivo and thereby provides new insights into developing a therapeutic strategy for mitochondria-related cardiovascular diseases such as heart failure.


Subject(s)
Cardiovascular Diseases , Heart Failure , Animals , Cardiomegaly/genetics , Mice , Mitochondria/genetics , Peroxiredoxin III/genetics , Protein Kinases
13.
Elife ; 102021 08 06.
Article in English | MEDLINE | ID: mdl-34355692

ABSTRACT

Amino-terminal acetylation is catalyzed by a set of N-terminal acetyltransferases (NATs). The NatA complex (including X-linked Naa10 and Naa15) is the major acetyltransferase, with 40-50% of all mammalian proteins being potential substrates. However, the overall role of amino-terminal acetylation on a whole-organism level is poorly understood, particularly in mammals. Male mice lacking Naa10 show no globally apparent in vivo amino-terminal acetylation impairment and do not exhibit complete embryonic lethality. Rather Naa10 nulls display increased neonatal lethality, and the majority of surviving undersized mutants exhibit a combination of hydrocephaly, cardiac defects, homeotic anterior transformation, piebaldism, and urogenital anomalies. Naa12 is a previously unannotated Naa10-like paralog with NAT activity that genetically compensates for Naa10. Mice deficient for Naa12 have no apparent phenotype, whereas mice deficient for Naa10 and Naa12 display embryonic lethality. The discovery of Naa12 adds to the currently known machinery involved in amino-terminal acetylation in mice.


Subject(s)
N-Terminal Acetyltransferase A/genetics , N-Terminal Acetyltransferase E/genetics , Acetylation , Animals , Female , Male , Mice , Mice, Knockout , N-Terminal Acetyltransferase A/deficiency , N-Terminal Acetyltransferase A/metabolism , N-Terminal Acetyltransferase E/deficiency , N-Terminal Acetyltransferase E/metabolism
14.
Materials (Basel) ; 13(19)2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33008052

ABSTRACT

More precast concrete structures have recently been constructed due to their many advantages when compared to conventional cast-in-place construction. Structural behavior at the joints between the precast segments can significantly affect the overall integrity, safety, and serviceability of the structure. In this study, therefore, the interface shear strength of high-strength precast members was investigated by performing push-off tests with the following variables: compressive strength of precast members, dry or wet joint, number and height of shear keys, joint width, filler type, curing temperature, and lateral compressive stress. The test results were analyzed to reveal the effect of each test variable on the joint shear strengths of the specimens. For instance, the failure loads were increased by 14-140%, depending on the lateral compressive stress, as the specified compressive strength of the precast members was increased from 80 to 150 MPa in the dry joints. The failure loads of the wet joints strongly depended on the strength of the filler rather than on that of the precast members and, as a result, the specimen with ultra-high-strength concrete filler was 46-48% stronger than those with high-strength mortar filler. The shear strengths of various joint types obtained from the test were further analyzed in comparison with the predictive equations of Japan Society of Civil Engineers (JSCE) and American Association of State Highway and Transportation Officials (AASHTO) with the aim of validating the appropriateness of these design provisions. In particular, an improved value of a coefficient in the JSCE equation is proposed to cover a range of compressive strengths in various precast members and filling materials.

15.
Redox Biol ; 37: 101760, 2020 10.
Article in English | MEDLINE | ID: mdl-33096425

ABSTRACT

Superoxide dismutase 1 (SOD1) binds copper and zinc ions and is one of three superoxide dismutases responsible for destroying free superoxide radicals in the body. Reactive oxygen species (ROS), including free superoxide radicals, play important roles in colitis. However, the role of SOD1 in oxidative stress under colitis remains unclear. Here, we examined the role of SOD1 in the DSS-induced mouse model of colitis. SOD1 deficiency resulted in severe oxidative stress with body weight loss, epithelial barrier disruption and decreased antioxidant enzyme activities. The levels of neutrophils, monocytes, pro-inflammatory CD11c+ macrophages and CD11b+CD103- dendritic cells (DCs) were increased, while anti-inflammatory CD206+ macrophages and CD11b-CD103+ DCs were decreased, in DSS-treated SOD1-knockout (KO) mice compared to DSS-treated wild-type mice. Furthermore, rescue of SOD activity in SOD1-KO mice by oral gavage of B. amyloliquefaciens SOD (BA SOD) significantly ameliorated enhanced DSS-induced colitis in these mice by suppressing p38-MAPK/NF-κB signaling, which can induce inflammation and apoptosis. Taken together, our results suggest that SOD1-mediated inhibitory responses play a crucial role in limiting the development of DSS-induced colitis, and that BA SOD is a promising candidate for treating colitis.


Subject(s)
Colitis , Oxidative Stress , Superoxide Dismutase-1 , Animals , Colitis/chemically induced , Colitis/genetics , Dextran Sulfate , Immunity , Mice , Mice, Inbred C57BL , Superoxide Dismutase-1/genetics
16.
Cell Rep ; 30(12): 4124-4136.e5, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32209473

ABSTRACT

CD137, a potent costimulatory receptor for CD8+ T cells, is expressed in various non-T cells, but little is known about its regulatory functions in these cells. In this study, we show that CD137 signaling, specifically in intestinal CD11b-CD103+ dendritic cells (DCs), restricts acute colitis progression. Mechanistically, CD137 engagement activates TAK1 and subsequently stimulates the AMPK-PGC-1α axis to enhance expression of the Aldh1a2 gene encoding the retinoic acid (RA) metabolizing enzyme RALDH2. RA can act on CD11b+CD103- DCs and induce SOCS3 expression, which, in turn, suppresses p38MAPK activation and interleukin-23 (IL-23) production. Administration of RA in DC-specific CD137-/- mice represses IL-23-producing CD11b+CD103- DCs and TH17 cells, indicating that RA is a major inhibitory effector molecule against intestinal CD11b+CD103- DCs. Additionally, the therapeutic effect of the anti-CD137 antibody is abrogated in DC-specific CD137-/- mice. Taken together, our results define a mechanism of paracrine immunoregulation operating between adjacent DC subsets in the intestine.


Subject(s)
Aldehyde Oxidoreductases/metabolism , Antigens, CD/metabolism , CD11b Antigen/metabolism , Colitis/pathology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Signal Transduction , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Acute Disease , Adenylate Kinase/metabolism , Animals , Apoptosis , Cell Differentiation , Colitis/immunology , Disease Susceptibility , Forkhead Transcription Factors/metabolism , Intestines/pathology , MAP Kinase Kinase Kinases/metabolism , Mice, Inbred C57BL , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/cytology , Tretinoin/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/deficiency
17.
Neuroimmunomodulation ; 16(6): 365-76, 2009.
Article in English | MEDLINE | ID: mdl-19609085

ABSTRACT

Neural progenitor cells (NPC) are self-renewing multipotent cells that generate neurons and glial cells in the brain. NPCs generate neurons and glia not only during development but also after neural injury. Recent studies have shown that endogenous NPCs are activated after brain injury and migrate toward damaged areas where astrocyte activation occurs. Considering the massive proliferation of astrocytes as well as the production of several kinds of cytoactive molecules after brain injury, such as NO, growth factors and cytokines, it is tempting to think that cytoactive molecules released by activated glial cells regulate neural progenitor differentiation and proliferation through inflammatory mediators. To test this hypothesis, we stimulated rat primary astrocytes with lipopolysaccharide (LPS) to induce the activation of astrocytes. After addition of the conditioned media from LPS-stimulated astrocytes to NPC culture, proliferation was examined by MTT assay and bromodeoxyuridine (BrdU) incorporation. The differentiation of NPC into neurons and astrocytes was examined by Western blot, ELISA and immunocytochemical staining with cell-type-specific markers. Conditioned media from LPS-stimulated astrocytes increased NPC proliferation as well as gliogenesis as compared with control conditioned media from astrocytes without LPS stimulation. In contrast, neurogenesis was decreased by LPS-conditioned media. To investigate the molecular mechanism mediating glial differentiation and proliferation of NPC by reactive astrocytes, we added inhibitors of the Erk and JNK pathways during LPS stimulation. These inhibitors - except for a p38 inhibitor - decreased NPC proliferation and glial differentiation. These results suggest that LPS stimulated astrocytes generate factors regulating NPC proliferation and gliogenesis via the Erk and JNK pathways.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Cell Proliferation , Cytokines/metabolism , Neurogenesis/physiology , Stem Cells/physiology , Animals , Astrocytes/cytology , Bromodeoxyuridine , Cell Communication/physiology , Cell Culture Techniques , Cells, Cultured , Culture Media, Conditioned/pharmacology , Enzyme Inhibitors/pharmacology , Indicators and Reagents , Inflammation Mediators , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Neurons/cytology , Neurons/physiology , Rats , Rats, Sprague-Dawley , Stem Cells/cytology , Tetrazolium Salts , Up-Regulation/physiology
18.
Exp Mol Med ; 51(1): 1-16, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30700695

ABSTRACT

Osteoclasts (OCs) are bone-resorbing cells that originate from hematopoietic stem cells and develop through the fusion of mononuclear myeloid precursors. Dysregulation of OC development causes bone disorders such as osteopetrosis, osteoporosis, and rheumatoid arthritis. Although the molecular mechanisms underlying osteoclastogenesis have been well established, the means by which OCs maintain their survival during OC development remain unknown. We found that Ninjurin1 (Ninj1) expression is dynamically regulated during osteoclastogenesis and that Ninj1-/- mice exhibit increased trabecular bone volume owing to impaired OC development. Ninj1 deficiency did not alter OC differentiation, transmigration, fusion, or actin ring formation but increased Caspase-9-dependent intrinsic apoptosis in prefusion OCs (preOCs). Overexpression of Ninj1 enhanced the survival of mouse macrophage/preOC RAW264.7 cells in osteoclastogenic culture, suggesting that Ninj1 is important for the survival of preOCs. Finally, analysis of publicly available microarray data sets revealed a potent correlation between high NINJ1 expression and destructive bone disorders in humans. Our data indicate that Ninj1 plays an important role in bone homeostasis by enhancing the survival of preOCs.


Subject(s)
Cancellous Bone/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Nerve Growth Factors/genetics , Osteoclasts/metabolism , Osteogenesis , Animals , Apoptosis , Cancellous Bone/growth & development , Cell Adhesion Molecules, Neuronal/metabolism , Cells, Cultured , Humans , Male , Mice , Nerve Growth Factors/metabolism , Osteoclasts/cytology , RAW 264.7 Cells
19.
Neurochem Res ; 33(11): 2324-34, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18493852

ABSTRACT

We investigated the effect of the cAMP system on lipopolysaccharide (LPS)-induced changes in the activity of matrix metalloproteinases (MMPs) and tissue plasminogen activator (tPA) in rat primary astrocytes. LPS stimulation increased MMP-9 and decreased tPA activity in rat primary astrocytes. Co-treatment with a cAMP analog, dibutyryl-cAMP (db-cAMP), or the cAMP elevating beta-adrenergic agonist, isoproterenol, concentration-dependently inhibited LPS-induced MMP-9 activity. In contrast, db-cAMP concentration-dependently increased tPA activity in both basal and LPS-stimulated rat primary astrocytes. To confirm the effect of cAMP on MMP-9 and tPA activity, we treated LPS-stimulated astrocytes with cAMP phosphodiesterase inhibitors, IBMX or rolipram, and they exhibited similar effects to db-cAMP, namely decreasing MMP-9 activity and increasing tPA activity. RT-PCR analysis of MMP-9 mRNA expression and MMP-9 promoter luciferase reporter assays revealed transcriptional upregulation by LPS stimulation and downregulation by db-cAMP. In contrast, the level of tPA mRNA expression was increased both by LPS and by cAMP treatment. Consistent with RT-PCR analysis, tPA promoter reporter assays showed increased activity by both LPS and cAMP stimulation. Interestingly, the level of mRNA encoding plasminogen activator inhibitor-1 (PAI-1) was increased by LPS stimulation and decreased back to control level after co-treatment with db-cAMP, suggesting that PAI-1 expression plays a major role in the regulation of tPA activity. To examine PKA involvement in the effects of db-cAMP on MMP-9 and tPA activity, we added the PKA inhibitors, H89 or rp-cAMP, along with db-cAMP, and they inhibited db-cAMP-mediated changes in tPA activity without affecting MMP-9 activity. These data suggest that cAMP differentially modulates MMP-9 and tPA activity through a mechanism related to PKA activation. The differential regulation of MMP-9 and tPA by the cAMP system may confer more sophisticated regulation of physiological processes, such as extracellular matrix remodeling and cell migration, by activated astrocytes.


Subject(s)
Astrocytes/drug effects , Cyclic AMP/metabolism , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 9/metabolism , Tissue Plasminogen Activator/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Base Sequence , Cells, Cultured , DNA Primers , Enzyme Activation , Promoter Regions, Genetic , RNA Processing, Post-Transcriptional , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Tissue Plasminogen Activator/genetics
20.
Autophagy ; 14(1): 120-133, 2018.
Article in English | MEDLINE | ID: mdl-28605287

ABSTRACT

Oxidative stress activates macroautophagy/autophagy and contributes to atherogenesis via lipophagic flux, a form of lipid removal by autophagy. However, it is not known exactly how endogenous antioxidant enzymes are involved in lipophagic flux. Here, we demonstrate that the antioxidant PRDX1 (peroxiredoxin 1) has a crucial role in the maintenance of lipophagic flux in macrophages. PRDX1 is more highly expressed than other antioxidant enzymes in monocytes and macrophages. We determined that Prdx1 deficiency induced excessive oxidative stress and impaired maintenance of autophagic flux in macrophages. Prdx1-deficient macrophages had higher intracellular cholesterol mass and lower cholesterol efflux compared with wild type. This perturbation in cholesterol homeostasis was due to impaired lipophagic cholesterol hydrolysis caused by excessive oxidative stress, resulting in the inhibition of free cholesterol formation and the reduction of NR1H3 (nuclear receptor subfamily 1, group H, member 3) activity. Notably, impairment of both lipophagic flux and cholesterol efflux was restored by the 2-Cys PRDX-mimics ebselen and gliotoxin. Consistent with this observation, apoe -/- mice transplanted with bone marrow from prdx1-/-apoe-/- mice had increased plaque formation compared with apoe-/- BM-transplanted recipients. This study reveals that PRDX1 is crucial to regulating lipophagic flux and maintaining macrophage cholesterol homeostasis against oxidative stress. We suggest that PRDX1-dependent control of oxidative stress may provide a strategy for treating atherosclerosis and autophagy-related human diseases.


Subject(s)
Autophagy , Cholesterol/metabolism , Macrophages/metabolism , Oxidative Stress , Peroxiredoxins/deficiency , Animals , Atherosclerosis/enzymology , Cells, Cultured , Humans , Liver X Receptors/metabolism , Mice , Mice, Knockout , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Peroxiredoxins/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL