Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(11): e0278220, 2022.
Article in English | MEDLINE | ID: mdl-36445856

ABSTRACT

Melanoma shows highly aggressive behavior (i.e., local invasion and metastasis). Matrix metalloprotease-3 (MMP-3), a zinc-dependent endopeptidase, degrades several extracellular substrates and contributes to local invasion by creating a microenvironment suitable for tumor development. Here, we report that interleukin-1ß (IL-1ß) triggers the MMP-3 expression in canine melanoma cells. The activity of MMP-3 in the culture supernatant was increased in IL-1ß-treated melanoma cells. IL-1ß time- and dose-dependently provoked the mRNA expression of MMP-3. IL-1ß induced the migration of melanoma cells; however, this migration was attenuated by UK356618, an MMP-3 inhibitor. When the cells were treated with the nuclear factor-κB (NF-κB) inhibitor TPCA-1, the inhibition of MMP-3 expression was observed. In IL-1ß-treated cells, the phosphorylation both of p65/RelA and p105 was detected, indicating NF-κB pathway activation. In p65/RelA-depleted melanoma cells, IL-1ß-mediated mRNA expression of MMP-3 was inhibited, whereas this reduction was not observed in p105-depleted cells. These findings suggest that MMP-3 expression in melanoma cells is regulated through IL-1ß-mediated p65/RelA activation, which is involved in melanoma cell migration.


Subject(s)
Matrix Metalloproteinase 3 , Melanoma , Animals , Dogs , Matrix Metalloproteinase 3/genetics , Interleukin-1beta/pharmacology , NF-kappa B , I-kappa B Proteins , RNA, Messenger , Tumor Microenvironment
2.
PLoS One ; 16(11): e0259489, 2021.
Article in English | MEDLINE | ID: mdl-34735542

ABSTRACT

In autoimmune diseases, fibroblasts produce and secrete various cytokines and act as sentinel immune cells during inflammatory states. However, the contribution of sentinel immune cells (i.e. dermal fibroblasts) in autoimmune diseases of the skin, such as atopic dermatitis, has been obscure. The pro-inflammatory cytokine interleukin 1ß (IL-1ß) induces the expression of chemokines, such as interleukin 8 (IL-8), in autoimmune diseases of the skin. IL-8 induces the activation and recruitment of innate immune cells such as neutrophils to the site of inflammation. IL-1ß-mediated induction of IL-8 expression is important for the pathogenesis of autoimmune diseases; however, the intracellular singling remains to be understood. To elucidate the mechanism of the onset of autoimmune diseases, we established a model for IL-1ß-induced dermatitis and investigated MAPK signaling pathways in IL-1ß-induced IL-8 expression. We also identified that a MAP3K Tpl2 acts as an upstream modulator of IL-1ß-induced ERK1/2 activation in dermal fibroblasts. We observed an increase in the expression of IL-8 mRNA and protein in cells treated with IL-1ß. ERK1/2 inhibitors significantly reduced IL-1ß-induced IL-8 expression, whereas the inhibitor for p38 MAPK or JNK had no effect. IL-1ß induced ERK1/2 phosphorylation, which was attenuated in the presence of an ERK1/2 inhibitor. IL-1ß failed to induce IL-8 expression in cells transfected with siRNA for ERK1, or ERK2. Notably, a Tpl2 inhibitor reduced IL-1ß-induced IL-8 expression and ERK1/2 phosphorylation. We confirmed that the silencing of Tpl2 in siRNA-transfected fibroblasts prevented both in IL-1ß-induced IL-8 expression and ERK1/2 phosphorylation. Taken together, our data indicate the importance of Tpl2 in the modulation of ERK1/2 signaling involved in the IL-1ß-induced development of autoimmune diseases affecting the dermal tissue, such as atopic dermatitis.


Subject(s)
Interleukin-1beta/pharmacology , Interleukin-8/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Blotting, Western , Cells, Cultured , Dogs , MAP Kinase Kinase Kinases/genetics , Male , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction
3.
PLoS One ; 16(2): e0243859, 2021.
Article in English | MEDLINE | ID: mdl-33539362

ABSTRACT

The rate of glucose uptake dramatically increases in cancer cells even in the presence of oxygen and fully functioning mitochondria. Cancer cells produce ATP by glycolysis rather than oxidative phosphorylation under aerobic conditions, a process termed as the "Warburg effect." In the present study, we treated canine melanoma cells with the glucose analog 2-deoxy-D-glucose (2-DG) and investigated its effect on cell growth. 2-DG attenuated cell growth in a time- and dose-dependent manner. Cell growth was also inhibited following treatment with the glucose transporter (GLUT) inhibitor WZB-117. The treatment of 2-DG and WZB-117 attenuated the glucose consumption, lactate secretion and glucose uptake of the cells. The mRNA expression of the subtypes of GLUT was examined and GLUT1 and GLUT3 were found to be expressed in melanoma cells. The growth, glucose consumption and lactate secretion of melanoma cells transfected with siRNAs of specific for GLUT1 and GLUT3 was suppressed. These findings suggest that glucose uptake via GLUT1 and GLUT3 plays a crucial role for the growth of canine melanoma cells.


Subject(s)
Cell Proliferation/physiology , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 3/metabolism , Melanoma/metabolism , Melanoma/pathology , Animals , Cell Line, Tumor , Dogs , Glucose/metabolism , Lactic Acid/metabolism , Male , RNA, Messenger/metabolism
4.
Front Immunol ; 11: 579266, 2020.
Article in English | MEDLINE | ID: mdl-33117381

ABSTRACT

The pro-inflammatory cytokine interleukin 1ß (IL-1ß) induces the synthesis of prostaglandin E2 by upregulating cyclooxygenase-2 (COX-2) in the synovial tissue of individuals with autoimmune diseases, such as rheumatoid arthritis (RA). IL-1ß-mediated stimulation of NF-κB and MAPK signaling is important for the pathogenesis of RA; however, crosstalk(s) between NF-κB and MAPK signaling remains to be understood. In this study, we established a model for IL-1ß-induced synovitis and investigated the role of NF-κB and MAPK signaling in synovitis. We observed an increase in the mRNA and protein levels of COX-2 and prostaglandin E2 release in cells treated with IL-1ß. NF-κB and ERK1/2 inhibitors significantly reduced IL-1ß-induced COX-2 expression. IL-1ß induced the phosphorylation of canonical NF-κB complex (p65 and p105) and degradation of IκBα. IL-1ß also induced ERK1/2 phosphorylation but did not affect the phosphorylation levels of p38 MAPK and JNK. IL-1ß failed to induce COX-2 expression in cells transfected with siRNA for p65, p105, ERK1, or ERK2. Notably, NF-κB inhibitors reduced IL-1ß-induced ERK1/2 phosphorylation; however, the ERK1/2 inhibitor had no effect on the phosphorylation of the canonical NF-κB complex. Although transcription and translation inhibitors had no effect on IL-1ß-induced ERK1/2 phosphorylation, the silencing of canonical NF-κB complex in siRNA-transfected fibroblasts prevented IL-1ß-induced phosphorylation of ERK1/2. Taken together, our data indicate the importance of the non-transcriptional/translational activity of canonical NF-κB in the activation of ERK1/2 signaling involved in the IL-1ß-induced development of autoimmune diseases affecting the synovial tissue, such as RA.


Subject(s)
Cyclooxygenase 2/metabolism , Fibroblasts/drug effects , Interleukin-1beta/toxicity , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , Synovial Membrane/drug effects , Synovitis/chemically induced , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Dogs , Enzyme Activation , Fibroblasts/enzymology , Fibroblasts/pathology , NF-kappa B/genetics , Phosphorylation , Signal Transduction , Synovial Membrane/enzymology , Synovial Membrane/pathology , Synovitis/enzymology , Synovitis/pathology
5.
PLoS One ; 15(3): e0229892, 2020.
Article in English | MEDLINE | ID: mdl-32231396

ABSTRACT

The specification of cell identity depends on the exposure of cells to sequences of bioactive ligands. All-trans retinoic acid (ATRA) affects neuronal development in the early stage, and it is involved in neuronal lineage reprogramming. We previously established a fibroblast-like dedifferentiated fat cells (DFATs) derived from highly homogeneous mature adipocytes, which are more suitable for the study of cellular reprogramming. Canine cognitive dysfunction is similar to human cognitive dysfunction, suggesting that dogs could be a pathological and pharmacological model for human neuronal diseases. However, the effect of ATRA on neuronal reprogramming in dogs has remained unclear. Therefore, in this study, we investigated the effect of ATRA on the neuronal reprogramming of canine DFATs. ATRA induced the expression of neuronal marker mRNA/protein. The neuron-like cells showed Ca2+ influx with depolarization (50 mM KCl; 84.75 ± 4.05%) and Na+ channel activation (50 µM veratridine; 96.02 ± 2.02%). Optical imaging of presynaptic terminal activity and detection of neurotransmitter release showed that the neuron-like cells exhibited the GABAergic neuronal property. Genome-wide RNA-sequencing analysis shows that the transcriptome profile of canine DFATs is effectively reprogrammed towards that of cortical interneuron lineage. Collectively, ATRA can produce functional GABAergic cortical interneuron-like cells from canine DFATs, exhibiting neuronal function with > 80% efficiency. We further demonstrated the contribution of JNK3 to ATRA-induced neuronal reprogramming in canine DFATs. In conclusion, the neuron-like cells from canine DFATs could be a powerful tool for translational research in cell transplantation therapy, in vitro disease modeling, and drug screening for neuronal diseases.


Subject(s)
Cell Dedifferentiation/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Tretinoin/pharmacology , Animals , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Dogs , Neurogenesis/genetics , RNA, Messenger/genetics , Synapses/drug effects , Synapses/genetics
6.
PLoS One ; 14(9): e0222869, 2019.
Article in English | MEDLINE | ID: mdl-31536594

ABSTRACT

Matrix metalloproteinases (MMPs) play a pivotal role in tissue remodeling by degrading the extracellular matrix (ECM) components. This mechanism is implicated in a variety of physiological and pathological cellular processes including wound healing. One of the key proteins involved in this process is the proinflammatory cytokine interleukin-1ß (IL-1ß, which induces the expression of MMP-3 mRNA and the secretion of MMP-3 protein by dermal fibroblasts. In this study, we first investigated the contribution of activating transcription factor 2 (ATF-2) to IL-1ß-induced MMP-3 expression in dermal fibroblasts. Our results showed that in cells transfected with ATF-2 siRNA or treated with the ATF-2 inhibitor SBI-0087702, IL-1ß-induced MMP-3 mRNA expression was reduced. We also demonstrated that IL-1ß stimulates the phosphorylation of ATF-2. These observations suggest that ATF-2 plays an important role in IL-1ß-induced MMP-3 expression. Next, we investigated the role of MAPK signaling in ATF-2 activation. In cells treated with the extracellular signal-regulated kinase (ERK) inhibitor FR180240, as well as in cells transfected with ERK1 and ERK2 siRNAs, IL-1ß-induced MMP-3 mRNA expression was reduced. In addition, we showed that IL-1ß induced the phosphorylation of ERK1/2. These observations suggest that ERK1 and ERK2 are involved in IL-1ß-induced MMP-3 expression. However, ERK1 and ERK2 do seem to play different roles. While the ERK inhibitor FR180204 inhibited IL-1ß-induced ATF-2 phosphorylation, only in cells transfected with ERK1 siRNA, but not ERK2 siRNA, IL-1ß-induced ATF-2 phosphorylation was reduced. These findings suggest that the ERK1/ATF-2 signaling axis contributes to IL-1ß-induced MMP-3 expression in dermal fibroblasts.


Subject(s)
Activating Transcription Factor 2/metabolism , Fibroblasts/drug effects , Interleukin-1beta/pharmacology , Matrix Metalloproteinase 3/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Activating Transcription Factor 2/genetics , Animals , Cells, Cultured , Dermis/cytology , Dogs , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Enzymologic/drug effects , Male , Matrix Metalloproteinase 3/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics
7.
PLoS One ; 14(7): e0220262, 2019.
Article in English | MEDLINE | ID: mdl-31344106

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine involved in the regulation of the immune response and inflammation. In this study, we investigated effect of the proinflammatory cytokine interleukin-1ß (IL-1ß) on IL-6 expression in canine dermal fibroblasts. IL-1ß induced IL-6 mRNA expression and protein release in a time- and dose-dependent manner. When cells were treated with inhibitors of mitogen-activated protein kinases (MAPKs), the extracellular signal-regulated kinase (ERK) inhibitor FR180240 inhibited IL-1ß-induced IL-6 mRNA expression, but not SP600125 or SKF86002, which are c-Jun N-terminal kinase (JNK) and p38 MAPK inhibitors, respectively. In cells treated with U0126, an inhibitor of MAPK/ERK kinase (MEK), which activates ERK, IL-1ß-induced IL-6 mRNA expression was also inhibited. IL-1ß stimulated ERK1/2 phosphorylation. In cells transfected with ERK1 and ERK2 isoform siRNAs, IL-1ß-induced IL-6 mRNA expression was reduced. These observations suggest that IL-1ß induces IL-6 expression via ERK1/2 signaling pathway in canine dermal fibroblasts.


Subject(s)
Dermis/drug effects , Fibroblasts/drug effects , Interleukin-1beta/pharmacology , Interleukin-6/genetics , Animals , Cells, Cultured , Dermis/cytology , Dermis/metabolism , Dogs , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Interleukin-6/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/genetics , Male , Signal Transduction/drug effects , Signal Transduction/genetics
8.
PLoS One ; 13(12): e0208955, 2018.
Article in English | MEDLINE | ID: mdl-30562372

ABSTRACT

Inflammatory and microenvironmental factors produced by cancer cells are thought to directly or indirectly promote cancer cell growth. Prostaglandins, including prostaglandin E2, have key roles as a microenvironment factor in influencing the development of tumors, and are produced by the rate limiting enzyme cyclooxygenase 2 (COX-2). In this study, we used canine melanoma cells treated with the proinflammatory cytokine interleukin 1ß (IL-1ß) and investigated the transcriptional factor nuclear factor-κB (NF-κB) signaling in IL-1ß-induced COX-2 expression. IL-1ß induced prostaglandin E2 release and COX-2 mRNA expression in a time- and dose-dependent manner. In the cells treated with the NF-κB inhibitors BAY11-7082 and TPC-1, IL-1ß-mediated prostaglandin E2 release and COX-2 mRNA expression were inhibited. IL-1ß also provoked phosphorylation of p65/RelA and p105/NF-κB1, which are members of the NF-κB families. The IL-1ß-induced phosphorylation of p65 and p105 was attenuated in the presence of both NF-κB inhibitors. In melanoma cells transfected with siRNA of p65 or p105, IL-1ß-mediated COX-2 mRNA expression was inhibited. These findings suggest that canonical activation of NF-κB signaling plays a crucial role for inflammatory states in melanoma cells.


Subject(s)
Antigens, Nuclear/genetics , Chromosomal Proteins, Non-Histone/genetics , Cyclooxygenase 2/genetics , Melanoma/genetics , Transcription Factor RelA/genetics , Animals , Disease Models, Animal , Dogs , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-1beta/genetics , Melanoma/pathology , Nitriles/pharmacology , Phosphorylation/drug effects , Signal Transduction/genetics , Sulfones/pharmacology
9.
Sci Rep ; 8(1): 8535, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29867151

ABSTRACT

The proinflammatory mediator bradykinin stimulated cyclooxygenase-2 (COX-2) expression and subsequently prostaglandin E2 synthesis in dermal fibroblasts. The involvement of B2 receptors and Gαq in the role of bradykinin was suggested by using pharmacological inhibitors. The PKC activator PMA stimulated COX-2 mRNA expression. Bradykinin failed to induce COX-2 mRNA expression in the presence of PKC inhibitors, whereas the effect of bradykinin was observed in the absence of extracellular Ca2+. Bradykinin-induced COX-2 mRNA expression was inhibited in cells transfected with PKCε siRNA. These observations suggest that the novel PKCε is concerned with bradykinin-induced COX-2 expression. Bradykinin-induced PKCε phosphorylation and COX-2 mRNA expression were inhibited by an inhibitor of 3-phosphoinositide-dependent protein kinase-1 (PDK-1), and bradykinin-induced PDK-1 phosphorylation was inhibited by phospholipase D (PLD) inhibitors, suggesting that PLD/PDK-1 pathway contributes to bradykinin-induced PKCε activation. Pharmacological and knockdown studies suggest that the extracellular signal-regulated kinase 1 (ERK1) MAPK signaling is involved in bradykinin-induced COX-2 expression. Bradykinin-induced ERK phosphorylation was attenuated in the cells pretreated with PKC inhibitors or transfected with PKCε siRNA. We observed the interaction between PKCε and ERK by co-immunoprecipitation experiments. These observations suggest that PKCε activation contributes to the regulation of ERK1 activation. Bradykinin stimulated the accumulation of phosphorylated ERK in the nuclear fraction, that was inhibited in the cells treated with PKC inhibitors or transfected with PKCε siRNA. Consequently, we concluded that bradykinin activates PKCε via the PLD/PDK-1 pathway, which subsequently induces activation and translocation of ERK1 into the nucleus, and contributes to COX-2 expression for prostaglandin E2 synthesis in dermal fibroblasts.


Subject(s)
Bradykinin/pharmacology , Cell Nucleus/enzymology , Cyclooxygenase 2/biosynthesis , Dermis/enzymology , Fibroblasts/enzymology , Mitogen-Activated Protein Kinase 3/metabolism , Protein Kinase C-epsilon/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Cell Line , Dermis/cytology , Dinoprostone/biosynthesis , Dogs , Fibroblasts/cytology , Gene Expression Regulation, Enzymologic/drug effects , MAP Kinase Signaling System/drug effects , Phosphorylation/drug effects
10.
PLoS One ; 12(8): e0182923, 2017.
Article in English | MEDLINE | ID: mdl-28806729

ABSTRACT

Tumor necrosis factor α (TNF-α) induces the expression and secretion of interleukin 8 (IL-8), which contributes to synovitis in rheumatoid arthritis (RA). To elucidate the mechanism of the onset of RA, we used synovial fibroblasts without autoimmune inflammatory diseases and investigated MAPK signaling pathways in TNF-α-induced IL-8 expression. Synovial fibroblasts isolated from healthy dogs were characterized by flow cytometry, which were positive for the fibroblast markers CD29, CD44, and CD90 but negative for the hematopoietic cell markers CD14, CD34, CD45, and HLA-DR. TNF-α stimulated the secretion and mRNA expression of IL-8 in a time- and dose-dependent manner. ERK and JNK inhibitors attenuated TNF-α-induced IL-8 expression and secretion. TNF-α induced the phosphorylation of ERK1/2 and JNK1/2. TNF-α-induced IL-8 expression was attenuated both in ERK2- and JNK1-knockdown cells. TNF-α-induced ERK1/2 or JNK1/2 was observed in ERK2- or JNK1-knockdown cells, respectively, showing that there is no crosstalk between ERK2 and JNK1 pathways. These observations indicate that the individual activation of ERK2 and JNK1 pathways contributes to TNF-α-induced IL-8 expression in synovial fibroblasts, which appears to be involved in the progress in RA.


Subject(s)
Fibroblasts/metabolism , Interleukin-8/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Synovial Membrane/cytology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Dogs , Enzyme Activation/drug effects , Fibroblasts/drug effects , Flow Cytometry , Humans , Interleukin-8/genetics , Isoenzymes/metabolism , MAP Kinase Signaling System/drug effects , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Sci Rep ; 7: 39914, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28054591

ABSTRACT

The proinflammatory cytokine interleukin 1ß (IL-1ß) induces prostaglandin E2 (PGE2) production via upregulation of cyclooxygenase-2 (COX-2) expression in synovial fibroblasts. This effect of IL-1ß is involved in osteoarthritis. We investigated MAPK signaling pathways in IL-1ß-induced COX-2 expression in feline synovial fibroblasts. In the presence of MAPK inhibitors, IL-1ß-induced COX-2 expression and PGE2 release were both attenuated. IL-1ß induced the phosphorylation of p38, JNK, MEK, and ERK1/2. A JNK inhibitor prevented not only JNK phosphorylation but also MEK and ERK1/2 phosphorylation in IL-1ß-stimulated cells, but MEK and ERK1/2 inhibitors had no effect on JNK phosphorylation. A p38 inhibitor prevented p38 phosphorylation, but had no effect on MEK, ERK1/2, and JNK phosphorylation. MEK, ERK1/2, and JNK inhibitors had no effect on p38 phosphorylation. We also observed that in IL-1ß-treated cells, phosphorylated MEK, ERK1/2, and JNK were co-precipitated with anti-phospho-MEK, ERK1/2, and JNK antibodies. The silencing of JNK1 in siRNA-transfected fibroblasts prevented IL-1ß to induce phosphorylation of MEK and ERK1/2 and COX-2 mRNA expression. These observations suggest that JNK1 phosphorylation is necessary for the activation of the MEK/ERK1/2 pathway and the subsequent COX-2 expression for PGE2 release, and p38 independently contributes to the IL-1ß effect in synovial fibroblasts.


Subject(s)
Cyclooxygenase 2/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblasts/physiology , Interleukin-1beta/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Signal Transduction , Animals , Cats , Cells, Cultured , Dinoprostone/metabolism
12.
PLoS One ; 11(11): e0166707, 2016.
Article in English | MEDLINE | ID: mdl-27851800

ABSTRACT

Acute kidney injury (AKI) is characterized by a sudden loss of renal function. Early recognition of AKI, especially in critically ill patients, is essential for adequate therapy. Currently, neutrophil gelatinase-associated lipocalin (NGAL) is considered to be an effective biomarker of AKI; however, the regulation of its expression and function in renal tubular cells remains unclear. In this study, we investigated the regulation of the expression and function of NGAL in IL-1ß-treated Madin-Darby canine kidney (MDCK) cells as a model of renal tubular cells. IL-1ß induced a disturbance in the localization of E-cadherin and zonaoccludin-1 (ZO-1). The transepithelial electrical resistance (TER) also decreased 5 days after IL-1ß treatment. IL-1ß induced NGAL mRNA expression and protein secretion in a time- and dose-dependent manner, which occurred faster than the decrease in TER. In the presence of ERK1/2 and p38 inhibitors, IL-1ß-induced NGAL mRNA expression and protein secretion were significantly attenuated. In the presence of recombinant NGAL, IL-1ß-induced disturbance in the localization of E-cadherin and ZO-1 was attenuated, and the decrease in TER was partially maintained. These results suggest that NGAL can be used as a biomarker for AKI and that it functions as a protector from AKI.


Subject(s)
Interleukin-1beta/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Lipocalin-2/metabolism , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Animals , Cadherins/metabolism , Dogs , Electric Impedance , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney Tubules/drug effects , Lipocalin-2/genetics , Madin Darby Canine Kidney Cells , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism , Zonula Occludens-1 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL