Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Publication year range
1.
J Clin Biochem Nutr ; 71(2): 103-111, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36213783

ABSTRACT

Vascular calcification is an important pathogenesis related to cardiovascular disease and high mortality rate in chronic kidney disease (CKD) patients. It has been well-known that hyper-phosphatemia induces osteochondrogenic transition of vascular smooth muscle cells (VSMCs) resulting ectopic calcification in aortic media, cardiac valve, and kidney. However, the detailed mechanism of the ectopic calcification has been not clarified yet. Here, we found that the co-localization of CYP27B1 with the calcified lesions of aorta and arteries in kidney of klotho mutant (kl/kl) mice, and then investigated the role of CYP27B1 in the mineralization of the VSMCs. Under high phosphate condition, overexpression of CYP27B1 induced calcification and osteocalcin mRNA expression in the VSMCs. Inversely, siRNA-CYP27B1 inhibited high phosphate-induced calcification of the VSMCs. We also found that the accumulated CYP27B1 protein was glycosylated in the kidney of kl/kl mice. Therefore, overexpression of CYP27B1-N310A and CYP27B1-T439A, which are a mutation for N-linked glycosylation site (N310A) and a mutation for O-linked glycosylation site (T439A) in CYP27B1, decreased calcium deposition and expression of RUNX2 induced by high phosphate medium in VSMCs compared with wild-type CYP27B1. These results suggest that extra-renal expression of glycosylated CYP27B1 would be required for ectopic calcification of VSMCs under hyperphosphatemia.

2.
Biochem J ; 477(4): 817-831, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32016357

ABSTRACT

Inorganic phosphate (Pi) homeostasis is regulated by intestinal absorption via type II sodium-dependent co-transporter (Npt2b) and by renal reabsorption via Npt2a and Npt2c. Although we previously reported that vitamin A-deficient (VAD) rats had increased urine Pi excretion through the decreased renal expression of Npt2a and Npt2c, the effect of vitamin A on the intestinal Npt2b expression remains unclear. In this study, we investigated the effects of treatment with all-trans retinoic acid (ATRA), a metabolite of vitamin A, on the Pi absorption and the Npt2b expression in the intestine of VAD rats, as well as and the underlying molecular mechanisms. In VAD rats, the intestinal Pi uptake activity and the expression of Npt2b were increased, but were reduced by the administration of ATRA. The transcriptional activity of reporter plasmid containing the promoter region of the rat Npt2b gene was reduced by ATRA in NIH3T3 cells overexpressing retinoic acid receptor (RAR) and retinoid X receptor (RXR). On the other hand, CCAAT/enhancer-binding proteins (C/EBP) induced transcriptional activity of the Npt2b gene. Knockdown of the C/EBP gene and a mutation analysis of the C/EBP responsible element in the Npt2b gene promoter indicated that C/EBP plays a pivotal role in the regulation of Npt2b gene transcriptional activity by ATRA. EMSA revealed that the RAR/RXR complex inhibits binding of C/EBP to Npt2b gene promoter. Together, these results suggest that ATRA may reduce the intestinal Pi uptake by preventing C/EBP activation of the intestinal Npt2b gene.


Subject(s)
Gene Expression Regulation/drug effects , Intestine, Small/metabolism , Kidney/metabolism , Promoter Regions, Genetic , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics , Transcription, Genetic/drug effects , Tretinoin/pharmacology , Animals , Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Hypophosphatemia, Familial/metabolism , Hypophosphatemia, Familial/pathology , Hypophosphatemia, Familial/prevention & control , Intestine, Small/drug effects , Kidney/drug effects , Male , Mice , NIH 3T3 Cells , Rats , Rats, Wistar , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIb/metabolism
3.
Biochem Biophys Res Commun ; 500(2): 275-282, 2018 06 02.
Article in English | MEDLINE | ID: mdl-29653103

ABSTRACT

The physiological activity of the steroid derived hormone vitamin D is regulated by several enzymatic steps. Both 25-hydroxy vitamin D3 1α-hydroxylase (CYP27B1) and 25-hydroxyvitamin D3 24-hydroxylase (CYP24A1) modulate blood levels of 1,25-dihydroxyvitamin D3, an activated form of vitamin D. We previously demonstrated that CYP27B1 expression was trans-activated by sterol regulatory element binding protein 1 (SREBP1), although whether SREBP1 also regulates CYP24A1 transcription was unclear. Here we investigated the ability of SREBP1 to affect CYP24A1 transcription. In a luciferase reporter assay, mouse and human CYP24A1 promoter activity was strongly activated by SREBP1 in opossum kidney proximal tubular cells (OK-P). Three putative SREs (pSREs) were found in the mouse Cyp24a1 gene promoter and the SREBP1 protein showed specific binding to the pSRE1 element in EMSAs. Site-directed mutagenesis of the pSRE1 element strongly decreased SREBP1-mediated Cyp24a1 gene transcription. Moreover, siRNA-mediated SREBP1 knock-down repressed CYP24A1 expression in human renal proximal tubular epithelial cells (HKC-8). In animal studies, mice given various doses of thyroid hormone (T3) showed dose-dependent reductions in renal Srebp1c and Cyp24a1 mRNA levels. Taken together, our results suggest that SREBP1 trans-activates CYP24A1 expression through SREBP binding elements present in the promoter.


Subject(s)
Gene Expression Regulation, Enzymologic , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcriptional Activation/genetics , Vitamin D3 24-Hydroxylase/genetics , Animals , Base Sequence , Cell Line , Humans , Mice , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription, Genetic
4.
J Vasc Surg ; 68(6S): 48S-59S.e1, 2018 12.
Article in English | MEDLINE | ID: mdl-29685509

ABSTRACT

OBJECTIVE: Osteoclastogenic activation of macrophages (OCG) occurs in human abdominal aortic aneurysms (AAAs) and in calcium chloride-induced degenerative AAAs in mice, which have increased matrix metalloproteinase activity. As the activity of OCG in dissecting aneurysms is not clear, we tested the hypothesis that OCG contributes to angiotensin II (Ang II)-induced dissecting aneurysm (Ang II-induced AAA) in apolipoprotein E knockout mice. METHODS: AAAs were produced in apolipoprotein E knockout mice via the administration of Ang II. Additionally, receptor activator of nuclear factor kB ligand (RANKL)-neutralizing antibody (5 mg/kg) was administered to one group of mice 7 days prior to Ang II infusion. Aneurysmal sections were probed for presence of RANKL and tartrate-resistant acid phosphatase via immunohistochemistry and immunofluorescence staining. Mouse aortas were also examined for RANKL and matrix metalloproteinase 9 expression via Western blot. In vitro murine vascular smooth muscle cells (MOVAS) and murine macrophages (RAW 264.7) were analyzed for the expression of osteogenic factors via Western blot, qPCR, and flow cytometry in response to Ang II or RANKL stimulation. The signaling pathway that mediates Ang II-induced RANKL expression in MOVAS cells was also investigated via application of TG101348, a Janus kinase 2 (JAK2) inhibitor, and Western blot analysis. RESULTS: Immunohistochemical staining of Ang II-induced AAA sections revealed OCG as evidenced by increased RANKL and tartrate-resistant acid phosphatase expression compared with control mice. Immunofluorescence staining of AAA sections revealed co-localization of vascular smooth muscle cells and RANKL, revealing vascular smooth muscle cells as one potential source of RANKL. Systemic administration of RANKL-neutralizing antibody suppressed Ang II-induced AAA, with significant reduction of the maximum diameter of the abdominal aorta compared with vehicle controls (1.5 ± 0.4 mm vs 2.2 ± 0.2 mm). Ang II (1 µM) treatment induced a significant increase in RANKL messenger RNA expression levels in MOVAS cells compared with the vehicle control (1.0 ± 0.2 vs 2.8 ± 0.2). The activities of JAK2 and signal transducer and activator of transcription 5 (STAT5) were also significantly increased by Ang II treatment. Inhibition of JAK2/STAT5 suppressed Ang II-induced RANKL expression, suggesting the involvement of the JAK2/STAT5 signaling pathway. CONCLUSIONS: OCG with increased RANKL expression was present in Ang II-induced AAA, and neutralization of RANKL suppressed AAA formation. As neutralization of RANKL has been used clinically to treat osteoporosis and other osteoclast-related diseases, additional study of the effectiveness of RANKL neutralization in AAA is warranted.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Aortic Dissection/metabolism , Cell Transdifferentiation , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Osteoclasts/metabolism , Osteogenesis , RANK Ligand/metabolism , Aortic Dissection/chemically induced , Aortic Dissection/pathology , Aortic Dissection/prevention & control , Angiotensin II , Animals , Antibodies, Neutralizing/pharmacology , Aorta, Abdominal/drug effects , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Cell Transdifferentiation/drug effects , Disease Models, Animal , Janus Kinase 2/metabolism , Macrophages/drug effects , Macrophages/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteogenesis/drug effects , RANK Ligand/antagonists & inhibitors , RANK Ligand/genetics , RANK Ligand/immunology , RAW 264.7 Cells , STAT5 Transcription Factor/metabolism , Signal Transduction
5.
Arterioscler Thromb Vasc Biol ; 36(9): 1962-71, 2016 09.
Article in English | MEDLINE | ID: mdl-27386936

ABSTRACT

OBJECTIVE: Arterial calcification is common and contributes to the pathogenesis of occlusive vascular disease. Similar to the dynamics of bone, it is a tightly controlled process that maintains a balance between osteogenesis and osteolysis. However, whether calcium homeostasis plays a role in the development of aneurysms has not been explored. We hypothesized that macrophages differentiate into osteoclasts in aneurysmal arteries and that protease byproducts contribute to aneurysm pathophysiology. APPROACH AND RESULTS: We performed histological and immunohistochemical analyses and showed that macrophages positive for several osteoclast markers, including tartrate acid phosphatase, occur in great numbers in the human aneurysmal aorta, but very few occur in the human stenotic aorta and none in the nondiseased human aorta. Moreover, in situ zymography showed elevated protease activity in these cells compared with undifferentiated macrophages. Tumor necrosis factor-α and calcium phosphate stimulated this osteoclastogenic differentiation process through nuclear factor-κB, mitogen-activated protein kinases, and intracellular calcium signaling but not the receptor activator of the nuclear factor-κB ligand. Inhibition of osteoclastogenic differentiation by bisphosphonate inhibits aneurysm development in a mouse model. CONCLUSIONS: These results suggest that differentiation of macrophages into osteoclasts contributes to the pathophysiology of aneurysmal disease.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Cell Transdifferentiation , Macrophages/metabolism , Osteoclasts/metabolism , Osteogenesis , Vascular Calcification/metabolism , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Calcium Chloride , Calcium Phosphates/pharmacology , Calcium Signaling , Cell Transdifferentiation/drug effects , Diphosphonates/pharmacology , Disease Models, Animal , Humans , Macrophages/drug effects , Macrophages/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Osteoclasts/pathology , Osteogenesis/drug effects , RAW 264.7 Cells , Tartrate-Resistant Acid Phosphatase/metabolism , Time Factors , Tumor Necrosis Factor-alpha/pharmacology , Vascular Calcification/pathology
6.
J Immunol ; 193(7): 3803-15, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25187658

ABSTRACT

Because ERK5 inhibits endothelial inflammation and dysfunction, activating ERK5 might be a novel approach to protecting vascular endothelial cells (ECs) against various pathological conditions of the blood vessel. We have identified small molecules that protect ECs via ERK5 activation and determined their contribution to preventing cardiac allograft rejection. Using high-throughput screening, we identified certain statins and antimalarial agents including chloroquine, hydroxychloroquine, and quinacrine as strong ERK5 activators. Pitavastatin enhanced ERK5 transcriptional activity and Kruppel-like factor-2 expression in cultured human and bovine ECs, but these effects were abolished by the depletion of ERK5. Chloroquine and hydroxychloroquine upregulated ERK5 kinase activity and inhibited VCAM-1 expression in an ERK5-dependent but MAPK/ERK kinase 5- and Kruppel-like factor 2/4-independent manner. Leukocyte rolling and vascular reactivity were used to evaluate endothelial function in vivo, and we found that EC-specific ERK5 knockout (ERK5-EKO) mice exhibited increased leukocyte rolling and impaired vascular reactivity, which could not be corrected by pitavastatin. The role of endothelial ERK5 in acute cardiac allograft rejection was also examined by heterotopic grafting of the heart obtained from either wild-type or ERK5-EKO mice into allomismatched recipient mice. A robust increase in both inflammatory gene expression and CD45-positive cell infiltration into the graft was observed. These tissue rejection responses were inhibited by pitavastatin in wild-type but not ERK5-EKO hearts. Our study has identified statins and antimalarial drugs as strong ERK5 activators and shown that ERK5 activation is preventive of endothelial inflammation and dysfunction and acute allograft rejection.


Subject(s)
Antimalarials/pharmacology , Endothelium, Vascular/immunology , Graft Rejection/drug therapy , Heart Transplantation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 7/immunology , Quinolines/pharmacology , Transcription, Genetic/drug effects , Allografts , Animals , Cattle , Endothelium, Vascular/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Human Umbilical Vein Endothelial Cells , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/immunology , Leukocyte Rolling/drug effects , Leukocyte Rolling/genetics , Leukocyte Rolling/immunology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 7/genetics , Transcription, Genetic/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
7.
Circulation ; 127(4): 486-99, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23243209

ABSTRACT

BACKGROUND: Diabetes mellitus is a major risk factor for cardiovascular mortality by increasing endothelial cell (EC) dysfunction and subsequently accelerating atherosclerosis. Extracellular-signal regulated kinase 5 (ERK5) is activated by steady laminar flow and regulates EC function by increasing endothelial nitric oxide synthase expression and inhibiting EC inflammation. However, the role and regulatory mechanisms of ERK5 in EC dysfunction and atherosclerosis are poorly understood. Here, we report the critical role of the p90 ribosomal S6 kinase (p90RSK)/ERK5 complex in EC dysfunction in diabetes mellitus and atherosclerosis. METHODS AND RESULTS: Inducible EC-specific ERK5 knockout (ERK5-EKO) mice showed increased leukocyte rolling and impaired vessel reactivity. To examine the role of endothelial ERK5 in atherosclerosis, we used inducible ERK5-EKO-LDLR(-/-) mice and observed increased plaque formation. When activated, p90RSK associated with ERK5, and this association inhibited ERK5 transcriptional activity and upregulated vascular cell adhesion molecule 1 expression. In addition, p90RSK directly phosphorylated ERK5 S496 and reduced endothelial nitric oxide synthase expression. p90RSK activity was increased in diabetic mouse vessels, and fluoromethyl ketone-methoxyethylamine, a specific p90RSK inhibitor, ameliorated EC-leukocyte recruitment and diminished vascular reactivity in diabetic mice. Interestingly, in ERK5-EKO mice, increased leukocyte rolling and impaired vessel reactivity were resistant to the beneficial effects of fluoromethyl ketone-methoxyethylamine, suggesting a critical role for endothelial ERK5 in mediating the salutary effects of fluoromethyl ketone-methoxyethylamine on endothelial dysfunction. Fluoromethyl ketone-methoxyethylamine also inhibited atherosclerosis formation in ApoE(-/-) mice. CONCLUSIONS: Our study highlights the importance of the p90RSK/ERK5 module as a critical mediator of EC dysfunction in diabetes mellitus and atherosclerosis formation, thus revealing a potential new target for therapeutic intervention.


Subject(s)
Atherosclerosis/physiopathology , Diabetic Angiopathies/physiopathology , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/metabolism , Drug Synergism , Glucose/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/pharmacology , Leukocyte Rolling/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Nitric Oxide Synthase Type III/metabolism , Oxidants/pharmacology , Phosphorylation/physiology , Rats , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors
8.
Am J Physiol Renal Physiol ; 306(7): F744-50, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24500689

ABSTRACT

The type IIa sodium-dependent phosphate cotransporter (Npt2a) plays a critical role in reabsorption of inorganic phosphate (Pi) by renal proximal tubular cells. Pi abnormalities during early stages of sepsis have been reported, but the mechanisms regulating Pi homeostasis during acute inflammation are poorly understood. We examined the regulation of Pi metabolism and renal Npt2a expression during lipopolysaccharide (LPS)-induced inflammation in mice. Dose-response and time-course studies with LPS showed significant increases of plasma Pi and intact parathyroid hormone (iPTH) levels and renal Pi excretion, while renal calcium excretion was significantly decreased. There was no difference in plasma 1,25-dihydroxyvitamin D levels, but the induction of plasma intact fibroblast growth factor 23 levels peaked 3 h after LPS treatment. Western blotting, immunostaining, and quantitative real-time PCR showed that LPS administration significantly decreased Npt2a protein expression in the brush border membrane (BBM) 3 h after injection, but there was no change in renal Npt2a mRNA levels. Moreover, tumor necrosis factor-α injection also increased plasma iPTH and decreased renal BBM Npt2a expression. Importantly, we revealed that parathyroidectomized rats had impaired renal Pi excretion and BBM Npt2a expression in response to LPS. These results suggest that the downregulation of Npt2a expression in renal BBM through induction of plasma iPTH levels alter Pi homeostasis during LPS-induced acute inflammation.


Subject(s)
Inflammation/metabolism , Kidney/metabolism , Lipopolysaccharides , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism , Acute Disease , Animals , Calcium/metabolism , Disease Models, Animal , Down-Regulation , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/blood , Inflammation/blood , Inflammation/chemically induced , Injections, Intraperitoneal , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Microvilli/metabolism , Parathyroid Hormone/blood , Parathyroidectomy , Phosphates/blood , Phosphates/urine , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Time Factors , Tumor Necrosis Factor-alpha/administration & dosage , Vitamin D/analogs & derivatives , Vitamin D/blood
9.
Circ Res ; 110(4): 536-50, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22267842

ABSTRACT

RATIONALE: Cardiomyocyte apoptosis is one of the key events in the development and progression of heart failure, and a crucial role for ICER (inducible cAMP early repressor) in this process has been previously reported. ERK5 is known to inhibit cardiac apoptosis after myocardial infarction (MI), especially in hyperglycemic states, via association with CHIP ubiquitin (Ub) ligase and subsequent upregulation of CHIP ligase activity, which induces ICER ubiquitination and subsequent protein degradation. The regulatory mechanism governing ERK5/CHIP interaction is unknown. OBJECTIVE: We previously demonstrated increased p90RSK activation in the diabetic heart. As a logical extension of this work, we now investigate whether p90RSK activation inhibits ERK5-mediated CHIP activation, and subsequently increases ICER levels and apoptosis. METHODS AND RESULTS: p90RSK activation inhibits ERK5/CHIP association and CHIP Ub ligase activity. p90RSK and CHIP share a common binding site in the ERK5 C-terminal domain (aa571-807). Overexpression of either p90RSK or an ERK5 fragment (aa571-807) inhibits ERK5/CHIP association, suggesting that p90RSK and CHIP competes for ERK5 binding and that p90RSK activation is critical for inhibiting ERK5/CHIP interaction. We also identified ERK5-S496 as being directly phosphorylated by p90RSK and demonstrated that an ERK5-S496A mutant significantly impairs Angiotensin II-mediated inhibition of CHIP activity and subsequent increase in ICER levels. In vivo, either cardiac-specific depletion of ERK5 or overexpression of p90RSK inhibits CHIP activity and accelerates cardiac apoptosis after MI-a phenomenon fully reversible by activating ERK5. CONCLUSIONS: These data suggest a role for p90RSK in inhibiting CHIP activity and promoting cardiac apoptosis through binding to and phosphorylation of ERK5-S496.


Subject(s)
Apoptosis , Diabetes Mellitus, Experimental/enzymology , Mitogen-Activated Protein Kinase 7/metabolism , Myocardial Infarction/enzymology , Myocytes, Cardiac/enzymology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Ubiquitin-Protein Ligases/metabolism , Angiotensin II/metabolism , Animals , Animals, Newborn , Binding Sites , Binding, Competitive , Cells, Cultured , Cyclic AMP Response Element Modulator/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Enzyme Activation , MAP Kinase Kinase 5/genetics , MAP Kinase Kinase 5/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitogen-Activated Protein Kinase 7/deficiency , Mitogen-Activated Protein Kinase 7/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction , Time Factors , Transfection , Ubiquitin-Protein Ligases/genetics
10.
Arterioscler Thromb Vasc Biol ; 33(2): 321-9, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23202365

ABSTRACT

OBJECTIVE: Protein inhibitor of activated signal transducer and activator of transcription-1 (PIAS1) is known to function as small ubiquitin-like modifier (SUMO) E3 ligase as well as transrepressor. The aim of the study is to elucidate the regulatory mechanisms for these 2 different functions, especially with respect to endothelial inflammation. METHODS AND RESULTS: The mitogen-activated protein kinase (MAPK)-activated protein kinase-2 is a proinflammatory kinase and phosphorylates PIAS1 at the Ser522 residue. Activation of MAPK-activated protein kinase-2 enhances p53-SUMOylation, but a PIAS1 phosphorylation mutant, PIAS1-S522A, abolished this p53-SUMOylation, suggesting a critical role for PIAS1-S522 phosphorylation in its SUMO ligase activity. Because nuclear p53 can inhibit Kruppel-like factor 2 promoter activity, we investigated the roles for PIAS1 phosphorylation and p53-SUMOylation in the Kruppel-like factor 2 and endothelial NO synthase expression. Both MAPK-activated protein kinase-2 and PIAS1 overexpression increased Kruppel-like factor 2 promoter activity and endothelial NO synthase expression, which were inhibited by expressing a p53-SUMOylation defective mutant, p53-K386R, and PIAS1-S522A. PIAS1-S522A also abolished the anti-inflammatory effect of wild-type PIAS1 in vitro and also in vivo, which was examined by leukocyte rolling in microvessels of skin grafts transduced by adenovirus encoding PIAS1-WT or - S522A mutant. CONCLUSIONS: Our study has identified a novel negative feedback regulatory pathway through which MAPK-activated protein kinase-2 limits endothelial inflammation via the PIAS1 S522 phosphorylation-mediated increase in PIAS1 transrepression and SUMO ligase activity.


Subject(s)
Endothelial Cells/enzymology , Inflammation/prevention & control , Intracellular Signaling Peptides and Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Protein Serine-Threonine Kinases/metabolism , Small Ubiquitin-Related Modifier Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cells, Cultured , Endothelial Cells/immunology , Enzyme Activation , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/immunology , Humans , Inflammation/enzymology , Inflammation/immunology , Inflammation Mediators/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Leukocyte Rolling , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , Protein Serine-Threonine Kinases/genetics , RNA Interference , Skin/blood supply , Skin Transplantation , Small Ubiquitin-Related Modifier Proteins/genetics , Sumoylation , Time Factors , Transfection , Transplantation, Autologous , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/genetics
11.
J Clin Biochem Nutr ; 54(2): 102-8, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24688219

ABSTRACT

Members of the fibroblast growth factor (FGF) 19 subfamily, including FGF23, FGF15/19, and FGF21, have a role as endocrine factors which influence the metabolism of inorganic phosphate (Pi) and vitamin D, bile acid, and energy. It has been reported that dietary Pi regulates circulating FGF23. In this study, the short-term effects of dietary Pi restriction on the expression of FGF19 subfamily members in mice were analyzed. An initial analysis confirmed plasma FGF23 levels positively correlated with the amount of dietary Pi. On the other hand, ileal Fgf15 gene expression, but not hepatic Fgf21 gene expression, was up-regulated by dietary Pi restriction. In addition, we observed the increase of plasma 1,25-dihydroxyvitamin D [1,25(OH)2D] levels by dietary Pi restriction, and the up-regulation of ileal Fgf15 mRNA expression by 1,25(OH)2D3 and vitamin D receptor (VDR). Importantly, dietary Pi restriction-induced Fgf15 gene expression was prevented in VDR-knockout mice. Furthermore, diurnal variations of plasma triglyceride concentrations and hepatic mRNA expression of the bile acid synthesis enzyme Cyp7a1 as one of Fgf15 negative target genes was influenced by dietary Pi restriction. These results suggest that dietary Pi restriction up-regulates ileal Fgf15 gene expression through 1,25(OH)2D3 and VDR, and may affect hepatic bile acid homeostasis.

12.
Biosci Microbiota Food Health ; 42(2): 131-137, 2023.
Article in English | MEDLINE | ID: mdl-37016689

ABSTRACT

The present study was conducted to elucidate the dietary effects of canna starch on the immune functions and intestinal luminal environment in mice. The amylose and resistant starch characteristics were determined for six types of starch, including edible canna. Canna starch was found to be higher in amylose and resistant starch compared with the other starches. BALB/c mice were fed 3.16% (low-canna group) and 6.32% (high-canna group) canna starch for 2 weeks, and then intestinal parameters were measured. Fecal IgA and mucin levels were markedly elevated by canna starch intake. IgA levels in serum and spleen lymphocytes were elevated by canna starch intake in the high-canna group, but not in the low-canna group. When the mice were fed canna starch, the cecum weight increased, and the pH in the cecum decreased. The high-canna group had significantly increased levels of Clostridium subcluster XIVa lactic acid, acetic acid, and n-butyric acid in the cecum compared with the control group. These results suggested that canna starch supplementation changed the intestinal microbiota and enhanced the intestinal immune and barrier functions and cecal organic acids in mice.

13.
J Oleo Sci ; 72(12): 1125-1131, 2023.
Article in English | MEDLINE | ID: mdl-38044136

ABSTRACT

Consumption of a high-fat diet (HFD) is associated with an increased risk of metabolic diseases, cancer, and neurological disorders, which are major global health concerns. In the present study, mice were fed a HFD containing 40% fat and 0.5% or 1.0% acylated steryl-ß-glucosides (ASG) and their gut microbiota was compared to that of mice fed with a low-fat diet (LFD). After 55 d, the epididymal fat weight was higher in the HFD and ASG groups than in the LFD group; however, the epididymal fat weight was lower in the ASG group than in the HFD group. The abundance of gut microbiota increased with HFD in obese micespecific Bacillota, but decreased when ASG was added to the HFD. The number of intestinal bacteria involved in the production of carcinogenic secondary bile acids was increased by the consumption of HFD, but decreased by the addition of ASG to HSD. This finding may indicate the gut bacteria-mediated health benefits of ASG.


Subject(s)
Diet, High-Fat , Gastrointestinal Microbiome , Mice , Animals , Diet, High-Fat/adverse effects , Glycosides , Sucrose , Obesity/microbiology , Glucosides
14.
Circ J ; 76(12): 2732-7, 2012.
Article in English | MEDLINE | ID: mdl-23117745

ABSTRACT

Arterial calcification is the result of the same highly organized processes as seen in bone, which rely on a delicate balance between osteoblasts and osteoclasts. Although previously understood as passive precipitation, evidence has accumulated to suggest that arterial calcification is the result of organized, regulated processes bearing many similarities to osteogenesis in bone, including the presence of subpopulations of arterial wall cells that retain osteoblastic lineage potential. These cells have the potential to form mineralized nodules and express osteoblast markers, including bone morphogenetic protein-2, osteocalcin, osteopontin, and alkaline phosphatase. By contrast, osteoclast-like cells mediate the catabolic process of mineral resorption. Recent data shows that cells positive for tartrate-resistant acid phosphatase, a major marker for osteoclasts, have been histologically identified in atherosclerotic lesions and are referred to as osteoclast-like cells. Evidence has accumulated to suggest that initial arterial calcification through passive precipitation of calcium phosphate initiates balanced mineralization regulated by osteoclast-like and osteoblast-like cells. Subsequently, various pathogenic conditions may trigger an imbalance between osteoblastogenesis and osteoclastogenesis, leading to either calcification in stenotic/occlusive disease or destruction of the extracellular matrix in aneurysmal disease. Further elucidation of these newly emerging concepts could lead to a novel therapeutic approach to arterial stenotic/occlusive disease and/or abdominal aortic aneurysm.


Subject(s)
Aortic Aneurysm, Abdominal/metabolism , Arterial Occlusive Diseases/metabolism , Bone Density , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis , Vascular Calcification/metabolism , Animals , Aortic Aneurysm, Abdominal/epidemiology , Aortic Aneurysm, Abdominal/pathology , Arterial Occlusive Diseases/epidemiology , Arterial Occlusive Diseases/pathology , Biomarkers/metabolism , Constriction, Pathologic , Diabetes Mellitus/epidemiology , Diabetes Mellitus/metabolism , Humans , Osteoblasts/pathology , Osteoclasts/pathology , Osteoporosis/epidemiology , Osteoporosis/metabolism , Prognosis , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/metabolism , Signal Transduction , Vascular Calcification/epidemiology , Vascular Calcification/pathology
15.
Biochem J ; 427(1): 161-9, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20088828

ABSTRACT

The type IIa renal sodium-dependent phosphate (Na/Pi) co-transporter Npt2a is implicated in the control of serum phosphate levels. It has been demonstrated previously that renal Npt2a protein and its mRNA expression are both up-regulated by the thyroid hormone T3 (3,3',5-tri-iodothyronine) in rats. However, it has never been established whether the induction was mediated by a direct effect of thyroid hormones on the Npt2a promoter. To address the role of Npt2a in T3-dependent regulation of phosphate homoeostasis and to identify the molecular mechanisms by which thyroid hormones modulate Npt2a gene expression, mice were rendered pharmacologically hypo- and hyper-thyroid. Hypothyroid mice showed low levels of serum phosphate and a marked decrease in renal Npt2a protein abundance. Importantly, we also showed that Npt2a-deficient mice had impaired serum phosphate responsiveness to T3 compared with wild-type mice. Promoter analysis with a luciferase assay revealed that the transcriptional activity of a reporter gene containing the Npt2a promoter and intron 1 was dependent upon TRs (thyroid hormone receptors) and specifically increased by T3 in renal cells. Deletion analysis and EMSAs (electrophoretic mobility-shift assays) determined that there were unique TREs (thyroid-hormone-responsive elements) within intron 1 of the Npt2a gene. These results suggest that Npt2a plays a critical role as a T3-target gene, to control phosphate homoeostasis, and that T3 transcriptionally activates the Npt2a gene via TRs in a renal cell-specific manner.


Subject(s)
Gene Expression Regulation , Kidney/metabolism , Phosphates/metabolism , Sodium-Phosphate Cotransporter Proteins, Type IIa/physiology , Transcription, Genetic/drug effects , Triiodothyronine/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Dogs , Electrophoretic Mobility Shift Assay , Female , HeLa Cells , Homeostasis , Humans , Kidney/cytology , Luciferases/metabolism , Male , Mice , Mice, Knockout , Promoter Regions, Genetic , Rats , Receptors, Thyroid Hormone/metabolism , Response Elements , Transcriptional Activation
16.
Biochem J ; 429(3): 583-92, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20507281

ABSTRACT

The type II sodium-dependent phosphate co-transporters Npt2a and Npt2c play critical roles in the reabsorption of Pi by renal proximal tubular cells. The vitamin A metabolite ATRA (all-trans-retinoic acid) is important for development, cell proliferation and differentiation, and bone formation. It has been reported that ATRA increases the rate of Pi transport in renal proximal tubular cells. However, the molecular mechanism is still unknown. In the present study, we observed the effects of a VAD (vitamin A-deficient) diet on Pi homoeostasis and the expression of Npt2a and Npt2c genes in rat kidney. There was no change in the plasma levels of Pi, but VAD rats significantly increased renal Pi excretion. Renal brush-border membrane Pi uptake activity and renal Npt2a and Npt2c expressions were significantly decreased in VAD rats. The transcriptional activity of a luciferase reporter plasmid containing the promoter region of human Npt2a and Npt2c genes was increased markedly by ATRA and a RAR (retinoic acid receptor)-specific analogue TTNPB {4-[E-2-(5,6,7,8-tetrahydro-5,5,8,8-tetra-methyl-2-naphtalenyl)-1-propenyl] benzoic acid} in renal proximal tubular cells overexpressing RARs and RXRs (retinoid X receptors). Furthermore, we identified RAREs (retinoic acid-response elements) in both gene promoters. Interestingly, the half-site sequences (5'-GGTTCA-3': -563 to -558) of 2c-RARE1 overlapped the vitamin D-responsive element in the human Npt2c gene and were functionally important motifs for transcriptional regulation of human Npt2c by ATRA and 1,25(OH)2D3 (1alpha,25-dihydroxyvitamin D3), in both independent or additive actions. In summary, we conclude that VAD induces hyperphosphaturia through the down-regulation of Npt2a and Npt2c gene expression in the kidney.


Subject(s)
Gene Expression Regulation/drug effects , Kidney/drug effects , Receptors, Drug/physiology , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Tretinoin/pharmacology , Animals , Blotting, Western , Diet , Electrophoretic Mobility Shift Assay , Gene Expression Regulation/physiology , Kidney/metabolism , Male , Polymerase Chain Reaction , Rats , Rats, Wistar , Transcription, Genetic/drug effects , Transcription, Genetic/physiology
17.
Commun Biol ; 3(1): 30, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949279

ABSTRACT

Communication between osteoblasts and osteoclasts plays a key role in bone metabolism. We describe here an unexpected role for matrix vesicles (MVs), which bud from bone-forming osteoblasts and have a well-established role in initiation of bone mineralization, in osteoclastogenesis. We show that the MV cargo miR-125b accumulates in the bone matrix, with increased accumulation in transgenic (Tg) mice overexpressing miR-125b in osteoblasts. Bone formation and osteoblasts in Tg mice are normal, but the number of bone-resorbing osteoclasts is reduced, leading to higher trabecular bone mass. miR-125b in the bone matrix targets and degrades Prdm1, a transcriptional repressor of anti-osteoclastogenic factors, in osteoclast precursors. Overexpressing miR-125b in osteoblasts abrogates bone loss in different mouse models. Our results show that the MV cargo miR-125b is a regulatory element of osteoblast-osteoclast communication, and that bone matrix provides extracellular storage of miR-125b that is functionally active in bone resorption.


Subject(s)
Bone Matrix/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism , Extracellular Vesicles/metabolism , MicroRNAs/genetics , Animals , Biological Transport , Biomarkers , Bone Resorption/pathology , Cell Communication , Gene Expression Regulation , Immunohistochemistry , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , Positive Regulatory Domain I-Binding Factor 1/genetics , RNA Interference , Signal Transduction
18.
Am J Physiol Endocrinol Metab ; 297(1): E76-84, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19435858

ABSTRACT

The mechanism by which replacement of some dietary carbohydrates with protein during weight loss favors lipid metabolism remains obscure. In this study, we investigated the effect of an energy-restricted, high-protein/low-carbohydrate diet on lipid metabolism in obese rats. High-sucrose-induced obese rats were assigned randomly to one of two energy-restricted dietary interventions: a carbohydrate-based control diet (CD) or a high-protein diet (HPD). Lean rats of the same age were assigned as normal control. There was significantly greater improvement in fatty liver and hypertriglyceridemia with the HPD diet relative to the CD diet. Expression of genes regulated by fibroblast growth factor-21 (FGF21) and involved in liver lipolysis and lipid utilitization, such as lipase and acyl-CoA oxidase, increased in obese rats fed the HPD. Furthermore, there was an inverse correlation between levels of FGF21 gene expression (regulated by glucagon/insulin balance) and increased triglyceride concentrations in liver from obese rats. Expression of hepatic stearoyl-CoA desaturase-1 (SCD1), regulated primarily by the dietary carbohydrate, was also markedly reduced in the HPD group (similar to plasma triglyceride levels in fasting animals) relative to the CD group. In conclusion, a hypocaloric high-protein diet improves fatty liver and hypertriglyceridemia effectively relative to a carbohydrate diet. The two cellular pathways at work behind these benefits include stimulation of hepatic lipolysis and lipid utilization mediated by FGF21 and reduction of hepatic VLDL-TG production by SCD1 regulation.


Subject(s)
Caloric Restriction , Dietary Proteins/therapeutic use , Fatty Liver/diet therapy , Hypertriglyceridemia/diet therapy , Obesity/diet therapy , Animals , Blood Glucose/metabolism , Cells, Cultured , Diet, Reducing/methods , Dietary Proteins/pharmacology , Fasting/blood , Fasting/metabolism , Fatty Liver/etiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Hypertriglyceridemia/etiology , Male , Obesity/chemically induced , Obesity/complications , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Sucrose
19.
J Nutr Sci Vitaminol (Tokyo) ; 63(1): 28-34, 2017.
Article in English | MEDLINE | ID: mdl-28367923

ABSTRACT

There have been reports that hyperglycemia suppresses osteoclast (OCL) differentiation, although the underlying mechanism is poorly understood. Here we demonstrated that high glucose suppresses OCL differentiation through activation of liver X receptor (LXR) ß, a recently reported glucose-sensing nuclear receptor. The effect of hyperglycemia on osteoclastogenesis was tested in RAW264.7 cells, a murine macrophage cell line. Cells were treated with receptor activator of NF-κB ligand (RANKL) under normoglycemic (5.5 mM glucose), normoglycemic with high osmotic pressure (5.5 mM glucose + 10.0 mM mannitol), and hyperglycemic (15.5 mM glucose) conditions. RANKL-induced osteoclastogenesis was significantly suppressed by high-glucose treatment. Mannitol treatment also significantly suppressed osteoclastogenesis, but the inhibitory effect was lower than for high-glucose treatment. The suppression of mRNA expression of Lxrß by RANKL was significantly restored by high glucose, but not mannitol. Additionally, the deactivation of Lxrß by siRNA attenuated high-glucose-induced suppression of osteoclastogenesis. Although further validation of the underlying pathway is necessary, targeting LXRß is a potential therapeutic approach to treating osteoporosis.


Subject(s)
Cell Differentiation/physiology , Hyperglycemia , Liver X Receptors/genetics , Osteoclasts/cytology , RANK Ligand/physiology , Animals , Gene Expression/drug effects , Glucose/pharmacology , Liver X Receptors/antagonists & inhibitors , Macrophages/cytology , Macrophages/drug effects , Mice , Osteogenesis/drug effects , RAW 264.7 Cells , RNA, Messenger/analysis , RNA, Small Interfering/pharmacology
20.
J Agric Food Chem ; 65(38): 8386-8391, 2017 Sep 27.
Article in English | MEDLINE | ID: mdl-28876919

ABSTRACT

We investigated whether Cirsium maritimum Makino can inhibit immunoglobulin-E-mediated allergic response in rat basophilic leukemia (RBL-2H3) cells and passive cutaneous anaphylaxis (PCA) in BALB/c mice. In vitro, the ethyl acetate extract of C. maritimum Makino (ECMM) significantly inhibited ß-hexosaminidase release and decreased intracellular Ca2+ levels in RBL-2H3 cells. Moreover, ECMM leaves more strongly suppressed the release of ß-hexosaminidase than ECMM flowers. ECMM leaves also significantly suppressed the PCA reaction in the murine model. High-performance liquid chromatography and 1H and 13C nuclear magnetic resonance indicated that cirsimaritin, a flavonoid, was concentrated in active fractions of the extract. Our findings suggest that ECMM leaves have a potential regulatory effect on allergic reactions that may be mediated by mast cells. Furthermore, cirsimaritin may be the active anti-allergic component in C. maritimum Makino.


Subject(s)
Anti-Allergic Agents/administration & dosage , Cirsium/chemistry , Hypersensitivity/drug therapy , Immunoglobulin E/immunology , Plant Extracts/administration & dosage , Animals , Antigens/immunology , Cell Line , Humans , Hypersensitivity/immunology , Male , Mast Cells/drug effects , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Passive Cutaneous Anaphylaxis/drug effects , Plant Leaves/chemistry , Rats , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/immunology
SELECTION OF CITATIONS
SEARCH DETAIL