Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Mol Clin Oncol ; 7(6): 976-980, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29285359

ABSTRACT

Mucinous tubular and spindle cell carcinoma (MTSCC) is rare in renal cell carcinoma (RCC) and usually demonstrates a low nuclear grade and a better prognosis compared with other RCCs. The authors present a case report of MTSCC containing an area of Fuhrman nuclear grade 3, in addition to an area with a micropapillary pattern. An 82-year-old man consulted a hospital due to macrohematuria, and a tumor in the right kidney was detected. The tumor was resected and histologically examined. The tumor consisted of various growth patterns: Elongated tubular structure, a papillary structure containing a micropapillary pattern and solid pattern with spindle cells. The tumor cells demonstrated Fuhrman nuclear grades 2 and 3. Invasion into the lymph vessel and metastasis into the regional lymph node were observed. Thus, the tumor was diagnosed as a high grade MTSCC. Five months following resection, a computed tomography scan suggested metastasis of the tumor into the para-aortic lymph nodes and liver, and the patient succumbed to brain metastasis. When MTSCC of kidney is observed, careful histological observation is important to avoid missing a high nuclear grade area.

2.
Oncol Lett ; 10(3): 1555-1560, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26622708

ABSTRACT

Angioimmunoblastic T cell lymphoma (AITL) is a peripheral T cell lymphoma, known to express CD3 and CD4, and, frequently, also CD10 and c-Maf-1. Hypergammaglobulinemia is not particularly rare in patients with AITL. However, AITL in conjunction with plasmacytosis in the peripheral blood is rare. The current report presents a case of CD10-negative AITL demonstrating leukemic change and plasmacytosis in the peripheral blood mimicking plasma cell leukemia. A 78-year-old male was admitted to hospital due to systemic lymph node enlargement, high serum IgG and IgA, and increased counts of plasmacytoid cells and lymphoid cells with atypical nuclei in the peripheral blood. Initially, plasma cell leukemia was suspected, due to the extreme increase in the number of plasma cells in the peripheral blood. However, the plasma cells did not show clonal expansion on examination by flow cytometry. Based on histological analyses, following a biopsy of an enlarged lymph node, the patient was diagnosed with AITL. This case suggests that when hypergammaglobulinemia and increases in B-lineage cells are observed, AITL should be considered in addition to disorders of B-lineage cells.

3.
Biochim Biophys Acta ; 1853(8): 1879-88, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25960397

ABSTRACT

Phosphatidylinositol 3-kinaseß (PI3Kß) plays a predominant role in integrin outside-in signaling and in platelet activation by GPVI engagement. We have shown that the tyrosine kinase Pyk2 mediates PI3Kß activation downstream of integrin αIIbß3, and promotes the phosphorylation of the PI3K-associated adaptor protein c-Cbl. In this study, we compared the functional correlation between Pyk2 and PI3Kß upon recruitment of the two main platelet collagen receptors, integrin α2ß1 and GPVI. PI3Kß-mediated phosphorylation of Akt was inhibited in Pyk2-deficient platelets adherent to monomeric collagen through integrin α2ß1, but occurred normally upon GPVI ligation. Integrin α2ß1 engagement led to Pyk2-independent association of c-Cbl with PI3K. However, c-Cbl was not phosphorylated in adherent platelets, and phosphorylation of Akt occurred normally in c-Cbl-deficient platelets, indicating that the c-Cbl is dispensable for Pyk2-mediated PI3Kß activation. Stimulation of platelets with CRP, a selective GPVI ligand, induced c-Cbl phosphorylation in the absence of Pyk2, but failed to promote its association with PI3K. Pyk2 activation was completely abrogated in PI3KßKD, but not in PI3KγKD platelets, and was strongly inhibited by Src kinases and phospholipase C inhibitors, and by BAPTA-AM. The absence of PI3Kß activity also hampered GPVI-induced tyrosine-phosphorylation and activation of PLCγ2, preventing intracellular Ca2+ increase and phosphorylation of pleckstrin. Moreover, GPVI-induced intracellular Ca2+ increase and pleckstrin phosphorylation were also strongly inhibited in human platelets treated with the PI3Kß inhibitor TGX-221. These results outline important differences in the regulation of PI3Kß by GPVI and integrin α2ß1 and suggest that inhibition of Pyk2 may target PI3Kß activation in a selective context of platelet stimulation.


Subject(s)
Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/physiology , Phosphatidylinositol 3-Kinases/metabolism , Platelet Membrane Glycoproteins/physiology , Proto-Oncogene Proteins c-cbl/physiology , Animals , Cells, Cultured , Enzyme Activation , Humans , Mice , Mice, Knockout , Signal Transduction
4.
Biochem J ; 469(2): 199-210, 2015 Jul 15.
Article in English | MEDLINE | ID: mdl-25967238

ABSTRACT

In blood platelets, stimulation of G protein-coupled receptors (GPCRs) by thrombin triggers the activation of Src family kinases (SFKs), resulting in the tyrosine-phosphorylation of multiple substrates, but the mechanism underlying this process is still poorly understood. In the present study, we show that the time-dependent protein-tyrosine phosphorylation triggered by thrombin in human or murine platelets was totally suppressed only upon concomitant chelation of intracellular Ca(2+) and inhibition of SFKs. Thrombin-induced activation of SFKs was regulated by intracellular Ca(2+) and accordingly the Ca(2+) ionophore A23187 was sufficient to stimulate SFKs. A23187 also triggered the phosphorylation and activation of the Ca(2+)-dependent focal adhesion kinase Pyk2 and Pyk2 activation by thrombin was Ca(2+)-dependent. Stimulation of SFKs by thrombin or A23187 was strongly reduced in platelets from Pyk2 knockout (KO) mice, as was the overall pattern of protein-tyrosine phosphorylation. By immunoprecipitation experiments, we demonstrate that Lyn and Fyn, but not Src, were activated by Pyk2. Inhibition of SFKs by PP2 also reduced the phosphorylation of Pyk2 in thrombin or A23187-stimulated platelets. Analysis of KO mice demonstrated that Fyn, but not Lyn, was required for complete Pyk2 phosphorylation by thrombin. Finally, PP2 reduced aggregation of murine platelets to a level comparable to that of Pyk2-deficient platelets, but did not have further effects in the absence of Pyk2. These results indicate that in thrombin-stimulated platelets, stimulation of Pyk2 by intracellular Ca(2+) initiates SFK activation, establishing a positive loop that reinforces the Pyk2/SFK axis and allows the subsequent massive tyrosine phosphorylation of multiple substrates required for platelet aggregation.


Subject(s)
Blood Platelets/enzymology , Calcium Signaling/drug effects , Focal Adhesion Kinase 2/metabolism , Hemostatics/pharmacology , Proto-Oncogene Proteins c-fyn/metabolism , Thrombin/pharmacology , src-Family Kinases/metabolism , Animals , Blood Platelets/cytology , Calcium Signaling/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Focal Adhesion Kinase 2/genetics , Humans , Mice , Mice, Knockout , Phosphorylation/drug effects , Phosphorylation/physiology , Platelet Activation/drug effects , Platelet Activation/genetics , Proto-Oncogene Proteins c-fyn/genetics , src-Family Kinases/genetics
5.
Immunobiology ; 220(9): 1059-66, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26004346

ABSTRACT

GVHD is a crucial mortality factor in allogeneic bone marrow transplantation (ABMT). In this paper, we show that dehydroxymethylepoxyquinomicin (DHMEQ), a novel inhibitor of nuclear factor-κB, suppresses GVHD, resulting in an improved mortality rate in a mouse ABMT model. Bone marrow cells from C57BL/6 mice (B6 mice) were transplanted into lethally irradiated BALB/c mice. Two weeks later, spleen cells from B6 mice were transplanted into the irradiated BALB/c mice. From one week after the injection of spleen cells, when the mice started to show GVHD, the mice were also injected intraperitoneally daily with DHMEQ or vehicle only (DMSO) for 4 weeks. By 80 days after the ABMT, 6/14 of the vehicle-injected mice (43%) had died because of GVHD, whereas all DHMEQ-injected mice survived this observation period and developed milder GVHD than the vehicle-injected mice. When regulatory T cells were reduced by the injection of anti-folate receptor 4 (FR4) antibody, the effects of DHMEQ were reduced. These findings suggest that administration of DHMEQ could become a new strategy for preventing fatalities from GVHD.


Subject(s)
Benzamides/therapeutic use , Bone Marrow Transplantation , Cyclohexanones/therapeutic use , Graft vs Host Disease/drug therapy , NF-kappa B/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , Cell Proliferation/drug effects , Cytokines/biosynthesis , Cytokines/genetics , Graft vs Host Disease/mortality , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/immunology , Spleen/cytology , Spleen/transplantation , Survival Rate , Transplantation, Homologous
6.
Clin Immunol ; 153(1): 64-72, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24721319

ABSTRACT

Kawasaki disease (KD) is a paediatric idiopathic vasculitis. In this study, on the basis of studies using an established animal model for KD, we report that mannose-binding lectin (MBL) is involved in the pathogenesis of the disease. KD-like experimental murine vasculitis was induced by intraperitoneally administering a Candida albicans water-soluble extract (CAWS). MBL-A gradually increased in the serum of the model mice treated with CAWS. Deposition of MBL-A and MBL-C was observed in the aortic root, including the coronary arteries, which is a predilection site in experimental vasculitis. Corresponding to the distribution patterns of MBLs, marked deposition of C3/C3-derived peptides was also observed. Regarding the self-reactivity of MBLs, we observed that MBLs interacted with core histones to activate the lectin pathway. These results suggest that some types of pathogens provoke the MBL-dependent complement pathway (lectin pathway) to cause and/or exacerbate KD-like vasculitis.


Subject(s)
Mannose-Binding Lectin/metabolism , Mucocutaneous Lymph Node Syndrome/immunology , Mucocutaneous Lymph Node Syndrome/metabolism , Animals , Antigens, Fungal/immunology , Aorta/metabolism , Aorta/pathology , Candida albicans/immunology , Complement Activation/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Disease Models, Animal , Histones/metabolism , Male , Mice , Signal Transduction
7.
Int J Gen Med ; 6: 575-9, 2013.
Article in English | MEDLINE | ID: mdl-23874118

ABSTRACT

For immunodeficient patients, fungi are life-threatening pathogens. In this paper, we present an autopsy case of combined zygomycosis and aspergillosis. A female in her 70s on chronic hemodialysis was admitted to a hospital suffering bloody sputum, dyspnea, and fever, probably due to perinuclear anti-neutrophil cytoplasmic antibody-related vasculitis. Antibiotics were administered and immunosuppressive therapy was started, resulting in an improvement in her condition. Pneumonia later developed, followed by pulmonary bleeding and intractable pneumothorax from which she ultimately died. On autopsy, the upper lobe of the left lung was found to have hemorrhagic necrosis and showed a large longitudinal fissure. Microscopically, Zygomycota were observed in both the lungs and heart, while Aspergillus was found in the middle lobe of the right lung. Zygomycosis, which usually has a poor prognosis, is assumed to have induced hemorrhagic infarction of the lungs, inducing pulmonary bleeding and necrosis, despite the use of lipid formulations of amphotericin B, which are effective medicines against Zygomycota.

8.
J Biol Chem ; 288(25): 18194-203, 2013 Jun 21.
Article in English | MEDLINE | ID: mdl-23640884

ABSTRACT

Proline-rich tyrosine kinase 2 (Pyk2) is activated by various agonists in platelets. We evaluated the signaling mechanism and the functional role of Pyk2 in platelets by using pharmacological inhibitors and Pyk2-deficient platelets. We found that platelet aggregation and secretion in response to 2-methylthio-ADP (2-MeSADP) and AYPGKF were diminished in the presence of Pyk2 inhibitors or in Pyk2-deficient platelets, suggesting that Pyk2 plays a positive regulatory role in platelet functional responses. It has been shown that ADP-, but not thrombin-induced thromboxane (TxA2) generation depends on integrin signaling. Unlike ADP, thrombin activates G12/13 pathways, and G12/13 pathways can substitute for integrin signaling for TxA2 generation. We found that Pyk2 was activated downstream of both G12/13 and integrin-mediated pathways, and both 2-MeSADP- and AYPGKF-induced TxA2 generation was significantly diminished in Pyk2-deficient platelets. In addition, TxA2 generation induced by co-stimulation of Gi and Gz pathways, which is dependent on integrin signaling, was inhibited by blocking Pyk2. Furthermore, inhibition of 2-MeSADP-induced TxA2 generation by fibrinogen receptor antagonist was not rescued by co-stimulation of G12/13 pathways in the presence of Pyk2 inhibitor. We conclude that Pyk2 is a common signaling effector downstream of both G12/13 and integrin αIIbß3 signaling, which contributes to thromboxane generation.


Subject(s)
Blood Platelets/metabolism , Focal Adhesion Kinase 2/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Thromboxane A2/biosynthesis , Adenosine Diphosphate/analogs & derivatives , Adenosine Diphosphate/pharmacology , Animals , Blood Platelets/drug effects , Blotting, Western , Cells, Cultured , Dose-Response Relationship, Drug , Focal Adhesion Kinase 2/antagonists & inhibitors , Focal Adhesion Kinase 2/genetics , Humans , Mice , Mice, Knockout , Oligopeptides/pharmacology , Phosphorylation/drug effects , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Thionucleotides/pharmacology , Time Factors , Tyrphostins/pharmacology
9.
Am J Nephrol ; 37(4): 302-9, 2013.
Article in English | MEDLINE | ID: mdl-23548793

ABSTRACT

BACKGROUND/AIMS: Minimal-change nephrotic syndrome (MCNS) is a kidney disease defined by selective proteinuria and hypoalbuminemia occurring in the absence of cellular glomerular infiltrates or immunoglobulin deposits. Recent observations suggest that nuclear factor κB (NF-κB) of podocyte is strongly associated with the development of proteinuria in MCNS. Dehydroxymethylepoxyquinomicin (DHMEQ) is a novel NF-κB inhibitor that potently inhibits DNA-binding activity of NF-κB, resulting in several therapeutic effects in various pathological conditions. We conducted this study to ask whether DHMEQ may ameliorate the nephrosis in mice induced by puromycin aminonucleoside (PAN), which is considered to be an animal model for MCNS. METHODS/RESULTS: Pretreatment with DHMEQ alleviated the proteinuria and reversed the serum abnormalities in mice nephrosis induced by 450 mg/kg of PAN. Increased serum interleukin-6 level in PAN-induced nephrosis was also completely suppressed by DHMEQ. Electron microscopic analyses of glo-meruli indicated that DHMEQ can inhibit the podocyte foot process effacement via blocking the translocation of podocyte NF-κB from cytoplasm to nucleus. CONCLUSIONS: These results suggest that DHMEQ can be a potential therapeutic agent for MCNS.


Subject(s)
Benzamides/administration & dosage , Cyclohexanones/administration & dosage , NF-kappa B/antagonists & inhibitors , Nephrosis/prevention & control , Puromycin Aminonucleoside/toxicity , Adenosine Deaminase/metabolism , Albuminuria/urine , Animals , Blood Proteins/analysis , Cholesterol/blood , Glycerolphosphate Dehydrogenase/metabolism , Interleukin-6/blood , Intracellular Signaling Peptides and Proteins/metabolism , Kidney/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , NF-kappa B/metabolism , Nephrosis/chemically induced , Nephrosis/metabolism , Nephrosis/pathology , Proteinuria/urine , Rats , Serum Albumin/analysis
10.
Blood ; 121(4): 648-57, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23175689

ABSTRACT

In the present study, we used a knockout murine model to analyze the contribution of the Ca(2+)-dependent focal adhesion kinase Pyk2 in platelet activation and thrombus formation in vivo. We found that Pyk2-knockout mice had a tail bleeding time that was slightly increased compared with their wild-type littermates. Moreover, in an in vivo model of femoral artery thrombosis, the time to arterial occlusion was significantly prolonged in mice lacking Pyk2. Pyk2-deficient mice were also significantly protected from collagen plus epinephrine-induced pulmonary thromboembolism. Ex vivo aggregation of Pyk2-deficient platelets was normal on stimulation of glycoprotein VI, but was significantly reduced in response to PAR4-activating peptide, low doses of thrombin, or U46619. Defective platelet aggregation was accompanied by impaired inside-out activation of integrin α(IIb)ß(3) and fibrinogen binding. Granule secretion was only slightly reduced in the absence of Pyk2, whereas a marked inhibition of thrombin-induced thromboxane A(2) production was observed, which was found to be responsible for the defective aggregation. Moreover, we have demonstrated that Pyk2 is implicated in the signaling pathway for cPLA(2) phosphorylation through p38 MAPK. The results of the present study show the importance of the focal adhesion kinase Pyk2 downstream of G-protein-coupled receptors in supporting platelet aggregation and thrombus formation.


Subject(s)
Focal Adhesion Kinase 2/genetics , Platelet Activation/genetics , Thrombin/metabolism , Thrombosis/genetics , Thrombosis/metabolism , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Group II Phospholipases A2/metabolism , Mice , Mice, Knockout , Phosphorylation , Platelet Activation/drug effects , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Signal Transduction , Thrombin/pharmacology , Thromboxane A2/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Ophthalmic Res ; 48(4): 199-207, 2012.
Article in English | MEDLINE | ID: mdl-22868688

ABSTRACT

PURPOSE: It has been reported that granulocyte colony-stimulating factor (G-CSF) provides neuroprotection in models in which neuronal cell death is induced. This research was designed to investigate the effects of G-CSF on neurodegeneration of the inner retinal layer in a rat model of ischemic reperfusion (I/R) injury. MATERIALS AND METHODS: Retinal ischemia was induced by increasing the intraocular pressure to 110 mm Hg for 45 min in the left eyes of the rats. A sham operation was carried out on the right eyes. G-CSF (100 µg/kg/day in 0.3 ml saline) or the same volume of saline was intraperitoneally injected just before the operation and continued for 4 consecutive days (a total of 5 consecutive days). Morphological examinations, including the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, were performed 7 days after I/R induction. The expression of phosphorylated AKT in the retina was examined by Western blot analysis and immunohistochemistry. RESULTS: Cell loss in the ganglion cell layer was more significantly reduced in the I/R-induced eyes of the G-CSF-injected rats than in the I/R-induced eyes of the saline-injected rats (20.3 vs. 6.6%). The inner retinal thickness ratios, such as the inner plexiform layer to the inner limiting membrane/outer nuclear layer and the inner nuclear layer/outer nuclear layer, were significantly better preserved in the I/R-induced eyes of the G-CSF-injected rats than in the I/R-induced eyes of the saline-injected rats. TUNEL assays showed fewer apoptotic cells in the retinal sections of the I/R-induced eyes of the G-CSF-injected rats. The phosphorylation of AKT (p-AKT/AKT) was upregulated in the retinas of the I/R-induced eyes of the G-CSF-injected rats. CONCLUSION: Our results demonstrated that systemic injection of G-CSF can protect retinal ganglion cells and inner retinal layers from I/R injury. The effects could be associated with the activation of AKT.


Subject(s)
Disease Models, Animal , Granulocyte Colony-Stimulating Factor/pharmacology , Neuroprotective Agents/pharmacology , Reperfusion Injury/prevention & control , Retinal Diseases/prevention & control , Retinal Ganglion Cells/drug effects , Animals , Apoptosis , Blotting, Western , Immunohistochemistry , In Situ Nick-End Labeling , Injections, Intraperitoneal , Leukocyte Count , Male , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Retinal Diseases/metabolism , Retinal Diseases/pathology , Retinal Ganglion Cells/pathology
12.
Biochem Biophys Res Commun ; 425(1): 39-44, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22820183

ABSTRACT

Vascular calcification is a major risk factor for the cardiovascular disease, yet its underlying molecular mechanisms remain to be elucidated. Recently, we identified that osteogenic signals via bone morphogenetic protein (BMP)-2 exerted by vascular smooth muscle cells (VSMCs) play a crucial role in the formation of atherosclerotic plaque calcification. Here we report a synergistic interaction between macrophages and VSMCs with respect to plaque calcification. Treatment with conditioned medium (CM) of macrophages dramatically enhanced BMP-2 expression in VSMCs, while it substantially reduced the expression of matrix Gla-protein (MGP) that inhibits the BMP-2 osteogenic signaling. As a result, macrophages significantly accelerated the osteoblastic differentiation of C2C12 cells induced by VSMC-CM. In contrast, macrophage-CM did not enhance the osteoblastic gene expressions in VSMCs, indicating that macrophages unlikely induced the osteoblastic trans-differentiation of VSMCs. We then examined the effect of recombinant TNF-α and IL-1ß on the VSMC-derived osteogenic signals. Similar to the macrophage-CM, both cytokines enhanced BMP-2 expression and reduced MGP expression in VSMCs. Nevertheless, only the neutralization of TNF-α but not IL-1ß attenuated the effect of macrophage-CM on the expression of these genes in VSMCs, due to the very low concentration of IL-1ß in the macrophage-CM. On the other hand, VSMCs significantly enhanced IL-1ß expression in macrophages, which might in turn accelerate the VSMC-mediated osteogenic signals. Together, we identified a unique role of macrophages in the formation of plaque calcification in coordination with VSMCs. This interaction between macrophages and VSMCs is a potential therapeutic target to treat and prevent the atherosclerotic plaque calcification.


Subject(s)
Macrophages/immunology , Muscle, Smooth, Vascular/immunology , Myocytes, Smooth Muscle/immunology , Osteogenesis/immunology , Plaque, Atherosclerotic/immunology , Vascular Calcification/immunology , Bone Morphogenetic Protein 2/biosynthesis , Calcium-Binding Proteins/biosynthesis , Cells, Cultured , Culture Media, Conditioned/pharmacology , Extracellular Matrix Proteins/biosynthesis , Humans , Interleukin-1beta/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Osteoblasts/immunology , Recombinant Proteins/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Vascular Calcification/drug therapy , Matrix Gla Protein
13.
J Mol Cell Cardiol ; 52(1): 175-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22044588

ABSTRACT

Inhibition of tumor suppressor p53 is cardioprotective against ischemic injury and provides resistance to subsequent cardiac remodeling. We investigated p53-mediated expansion of ischemic damage with a focus on mitochondrial integrity in association with autophagy and apoptosis. p53(-/-) heart showed that autophagic flux was promoted under ischemia without a change in cardiac tissue ATP content. Electron micrographs revealed that ischemic border zone in p53(-/-) mice had 5-fold greater numbers of autophagic vacuoles containing mitochondria, indicating the occurrence of mitophagy, with an apparent reduction of abnormal mitochondria compared with those in WT mice. Analysis of autophagic mediators acting downstream of p53 revealed that TIGAR (TP53-induced glycolysis and apoptosis regulator) was exclusively up-regulated in ischemic myocardium. TIGAR(-/-) mice exhibited the promotion of mitophagy followed by decrease of abnormal mitochondria and resistance to ischemic injury, consistent with the phenotype of p53(-/-) mice. In p53(-/-) and TIGAR(-/-) ischemic myocardium, ROS production was elevated and followed by Bnip3 activation which is an initiator of mitophagy. Furthermore, the activation of Bnip3 and mitophagy due to p53/TIGAR inhibition were reversed with antioxidant N-acetyl-cysteine, indicating that this adaptive response requires ROS signal. Inhibition of mitophagy using chloroquine in p53(-/-) or TIGAR(-/-) mice exacerbated accumulation of damaged mitochondria to the level of wild-type mice and attenuated cardioprotective action. These findings indicate that p53/TIGAR-mediated inhibition of myocyte mitophagy is responsible for impairment of mitochondrial integrity and subsequent apoptosis, the process of which is closely involved in p53-mediated ventricular remodeling after myocardial infarction.


Subject(s)
Myocardial Ischemia/metabolism , Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins , Autophagy/genetics , Gene Expression Regulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria, Heart/metabolism , Mitochondria, Heart/pathology , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/metabolism , Myocardial Ischemia/genetics , Oxidative Stress , Phosphoric Monoester Hydrolases , Proteins/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Ventricular Remodeling/genetics
14.
Blood ; 119(3): 847-56, 2012 Jan 19.
Article in English | MEDLINE | ID: mdl-22106345

ABSTRACT

Integrin α2ß1-mediated adhesion of human platelets to monomeric type I collagen or to the GFOGER peptide caused a time-dependent activation of PI3K and Akt phosphorylation. This process was abrogated by pharmacologic inhibition of PI3Kß, but not of PI3Kγ or PI3Kα. Moreover, Akt phosphorylation was undetectable in murine platelets expressing a kinase-dead mutant of PI3Kß (PI3Kß(KD)), but occurred normally in PI3Kγ(KD) platelets. Integrin α2ß1 failed to stimulate PI3Kß in platelets from phospholipase Cγ2 (PLCγ2)-knockout mice, and we found that intracellular Ca(2+) linked PLCγ2 to PI3Kß activation. Integrin α2ß1 also caused a time-dependent stimulation of the focal kinase Pyk2 downstream of PLCγ2 and intracellular Ca(2+). Whereas activation of Pyk2 occurred normally in PI3Kß(KD) platelets, stimulation of PI3Kß was strongly reduced in Pyk2-knockout mice. Neither Pyk2 nor PI3Kß was required for α2ß1-mediated adhesion and spreading. However, activation of Rap1b and inside-out stimulation of integrin αIIbß3 were reduced after inhibition of PI3Kß and were significantly impaired in Pyk2-deficient platelets. Finally, both PI3Kß and Pyk2 significantly contributed to thrombus formation under flow. These results demonstrate that Pyk2 regulates PI3Kß downstream of integrin α2ß1, and document a novel role for Pyk2 and PI3Kß in integrin α2ß1 promoted inside-out activation of integrin αIIbß3 and thrombus formation.


Subject(s)
Blood Platelets/metabolism , Focal Adhesion Kinase 2/physiology , Integrin alpha2beta1/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Platelet Adhesiveness , Proto-Oncogene Proteins c-akt/metabolism , Animals , Calcium/metabolism , Collagen/metabolism , Fibrinogen/metabolism , Humans , Immunoblotting , Mice , Mice, Knockout , Phosphorylation , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction
15.
Circ Heart Fail ; 5(1): 106-15, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22075967

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy is characterized by energetic dysregulation caused by glucotoxicity, lipotoxicity, and mitochondrial alterations. p53 and its downstream mitochondrial assembly protein, synthesis of cytochrome c oxidase 2 (SCO2), are important regulators of mitochondrial respiration, whereas the involvement in diabetic cardiomyopathy remains to be determined. METHODS AND RESULTS: The role of p53 and SCO2 in energy metabolism was examined in both type I (streptozotocin [STZ] administration) and type II diabetic (db/db) mice. Cardiac expressions of p53 and SCO2 in 4-week STZ diabetic mice were upregulated (185% and 152% versus controls, respectively, P<0.01), with a marked decrease in cardiac performance. Mitochondrial oxygen consumption was increased (136% versus control, P<0.01) in parallel with augmentation of mitochondrial cytochrome c oxidase (complex IV) activity. Reactive oxygen species (ROS)-damaged myocytes and lipid accumulation were increased in association with membrane-localization of fatty acid translocase protein FAT/CD36. Antioxidant tempol reduced the increased expressions of p53 and SCO2 in STZ-diabetic hearts and normalized alterations in mitochondrial oxygen consumption, lipid accumulation, and cardiac dysfunction. Similar results were observed in db/db mice, whereas in p53-deficient or SCO2-deficient diabetic mice, the cardiac and metabolic abnormalities were prevented. Overexpression of SCO2 in cardiac myocytes increased mitochondrial ROS and fatty acid accumulation, whereas knockdown of SCO2 ameliorated them. CONCLUSIONS: Myocardial p53/SCO2 signal is activated by diabetes-mediated ROS generation to increase mitochondrial oxygen consumption, resulting in excessive generation of mitochondria-derived ROS and lipid accumulation in association with cardiac dysfunction.


Subject(s)
Diabetes Complications/complications , Diabetes Mellitus, Experimental/complications , Diabetic Cardiomyopathies/etiology , Lipid Metabolism/physiology , Mitochondria, Heart/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , CD36 Antigens/metabolism , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetic Cardiomyopathies/metabolism , Disease Models, Animal , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Fatty Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Molecular Chaperones , Myocytes, Cardiac/metabolism , Oxygen Consumption/physiology , Streptozocin/adverse effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
Kidney Int ; 81(5): 449-57, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22157654

ABSTRACT

Unilateral ureteral obstruction is a well-established experimental model of progressive renal fibrosis. We tested whether mechanical stretch and subsequent renal tubular distension might lead to renal fibrosis by first studying renal tubular epithelial cells in culture. We found that mechanical stretch induced reactive oxygen species that in turn activated the cytoplasmic proline-rich tyrosine kinase-2 (Pyk2). This kinase is abundantly expressed in tubular epithelial cells where it is activated by several stimuli. Using mice with deletion of Pyk2 we found that the expression of transforming growth factor-ß1 induced by mechanical stretch in renal tubular epithelial cells was significantly reduced. The expression of connective tissue growth factor was also reduced in the Pyk2(-/-) mice. We also found that expression of connective tissue growth factor was independent of transforming growth factor-ß1, but dependent on the Rho-associated coiled-coil forming protein kinase pathway. Thus, Pyk2 may be an important initiating factor in renal fibrosis and might be a new therapeutic target for ameliorating renal fibrosis.


Subject(s)
Connective Tissue Growth Factor/metabolism , Focal Adhesion Kinase 2/metabolism , Kidney Tubules/metabolism , Kidney/metabolism , Kidney/pathology , Stress, Mechanical , Animals , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibrosis , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/genetics , Kidney Tubules/pathology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Phosphorylation , Reactive Oxygen Species/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Ureteral Obstruction/complications
17.
Hypertension ; 58(2): 254-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21730299

ABSTRACT

Ageing is an important risk factor for ischemic cardiovascular diseases, although its underlying molecular mechanisms remain to be elucidated. Here, we report a crucial role of Bcl-2 in the impaired angiogenic functions in senescent endothelial cells (ECs) by modulating the mitochondrial redox state. Cellular senescence impaired angiogenic functions in ECs without attenuating the mitogen-activated protein kinase or Akt signaling, and vascular endothelial growth factor receptor 2 or Tie-2 expressions. We identified that Bcl-2 expression was markedly reduced in 3 independent models for senescent ECs, and pharmacological inhibition, as well as small interfering RNA-mediated gene silencing of Bcl-2, significantly impaired the angiogenic functions in young ECs. Bcl-2 has an antioxidative role by locating the glutathione at mitochondria, and we found that mitochondrial oxidative stress was significantly augmented in senescent ECs, in association with reduced mitochondria-associated glutathione. Transfection of Bcl-2 in senescent ECs significantly reduced the mitochondrial oxidative stress, restored the mitochondrial membrane potential, and improved the angiogenic capacity. Furthermore, gene transfer of Bcl-2 using adenovirus significantly improved the in vivo angiogenesis in the Matrigel plugs implanted into aged mice, whereas the Bcl-2 inhibitor reduced the angiogenesis in the Matrigel plugs implanted into young mice. Together, Bcl-2 plays a crucial role in the regulation of the mitochondrial redox state in ECs, and, thus, loss of Bcl-2 during the senescence exacerbates the impaired angiogenesis by augmenting the mitochondrial oxidative stress.


Subject(s)
Cellular Senescence/physiology , Endothelial Cells/metabolism , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cells, Cultured , Endothelial Cells/cytology , Humans , Membrane Potential, Mitochondrial , Mice , Mitogen-Activated Protein Kinases/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidation-Reduction , Oxidative Stress , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Umbilical Veins/cytology , Umbilical Veins/metabolism
18.
Proc Natl Acad Sci U S A ; 108(23): 9472-7, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21593423

ABSTRACT

Endothelial and endothelial progenitor cells (ECs and EPCs) play a fundamental role in angiogenesis that is essential for numerous physiological and pathological processes. The phosphatase and tensin homolog (PTEN)/ phosphoinositide 3-kinase (PI3K) pathway has been implicated in angiogenesis, but the mechanism in the regulation of this pathway in ECs and EPCs is poorly understood. Here we show that ARIA (apoptosis regulator through modulating IAP expression), a transmembrane protein that we recently identified, regulates the PTEN/PI3K pathway in ECs and EPCs and controls developmental and postnatal angiogenesis in vivo. We found that ARIA is abundantly expressed in EPCs and regulates their angiogenic functions by modulating PI3K/Akt/endothelial nitric oxide synthase (eNOS) signaling. Genetic deletion of ARIA caused nonfatal bleeding during embryogenesis, in association with increased small vessel density and altered expression of various vascular growth factors including angiopoietins and VEGF receptors. Postnatal neovascularization induced by critical limb ischemia was substantially enhanced in ARIA-null mice, in conjunction with more bone marrow (BM)-derived ECs detected in ischemic muscles. Administration of PI3K or NO synthase inhibitor completely abolished the enhanced neovascularization in ARIA(-/-) mice. Mechanistically, we identified that ARIA interacts with PTEN at the intracellular domain independently of the PTEN phosphorylation in its C-terminal tail. Overexpressed ARIA increased PTEN in the membrane fraction, whereas ARIA-silencing reduced the membrane-associated PTEN, resulting in modified PI3K/Akt signaling. Taken together, our findings establish a previously undescribed mode of regulation of the PTEN/PI3K/Akt pathway by ARIA, and reveal a unique mechanism in the control of angiogenesis. These functions of ARIA might offer a unique therapeutic potential.


Subject(s)
Endothelial Cells/metabolism , Neuregulin-1/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stem Cells/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blood Vessels/embryology , Blood Vessels/growth & development , Blood Vessels/metabolism , CHO Cells , Cell Line , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Cricetulus , Endothelial Cells/cytology , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Knockout , Mutation , Neuregulin-1/genetics , Nitric Oxide Synthase Type III/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Protein Binding , RNA Interference , Signal Transduction , Stem Cells/cytology
19.
Arterioscler Thromb Vasc Biol ; 31(5): 1084-92, 2011 May.
Article in English | MEDLINE | ID: mdl-21372295

ABSTRACT

OBJECTIVE: Reactive oxygen species (ROS) are involved in the initial process of atherosclerosis, whereas it remains to be determined how atherogenic stimulus causes ROS-mediated proinflammatory reactions. Here, we focused on proline-rich tyrosine kinase (PYK2)-mediated ROS generation and examined how atherogenic stimulus causes early proinflammatory reactions. METHODS AND RESULTS: PYK2-deficient (knockout [KO]) (PYK2-KO) mice were crossbred with apolipoprotein E (ApoE)-deficient (PYK2-KO/ApoE-KO) mice. PYK2-KO/ApoE-KO mice and endothelial cells (EC) were used for the study. Aortic atherogenic lesions in PYK2-KO/ApoE-KO mice were markedly decreased (55% versus ApoE-KO) after 8 weeks of a Western diet. Aortic PYK2 was activated as early as 7 days after the Western diet, when inflammatory cells were not yet activated. Addition of the proatherogenic oxidized phospholipid lysophosphatidylcholine caused activation of endothelial PYK2. Lysophosphatidylcholine-activated PYK2 induced NADPH oxidase-mediated ROS generation and ROS-mediated synthesis of tumor necrosis factor-α (TNFα), vascular cell adhesion molecule-1 (VCAM-1), monocyte chemotactic protein-1 (MCP-1), and p21Cip1/Ets-1. Neutralizing anti-TNFα antibody or knockdown of p21Cip1/Ets-1 system blocked the induction of VCAM-1 and MCP-1. PYK2 deficiency abolished these ROS-mediated proinflammatory reactions. Further analysis revealed that PYK2/ROS-mediated p21Cip1/Ets-1 activation upregulated the transcription of the MCP-1 gene in collaboration with p300 transcription coactivator. CONCLUSIONS: PYK2 is a key tyrosine kinase activated by high cholesterol exposure, which causes ROS-mediated TNFα release and induces TNFα-dependent expression of proinflammatory molecules via the p21Cip1/Ets-1/p300 transcription system.


Subject(s)
Aortic Diseases/enzymology , Atherosclerosis/enzymology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Endothelial Cells/enzymology , Focal Adhesion Kinase 2/metabolism , Inflammation Mediators/metabolism , Proto-Oncogene Protein c-ets-1/metabolism , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Aortic Diseases/genetics , Aortic Diseases/pathology , Aortic Diseases/prevention & control , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Bone Marrow Transplantation , Cells, Cultured , Chemokine CCL2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Disease Models, Animal , Endothelial Cells/pathology , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/genetics , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Lipoproteins, LDL/metabolism , Lysophosphatidylcholines/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/metabolism , Proto-Oncogene Protein c-ets-1/genetics , RNA Interference , Signal Transduction , Time Factors , Transcriptional Activation , Transfection , Vascular Cell Adhesion Molecule-1/metabolism , p300-CBP Transcription Factors/genetics
20.
Am J Physiol Heart Circ Physiol ; 300(1): H154-61, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20952669

ABSTRACT

Members of the fibroblast growth factor (FGF) family have been clinically applied to the treatment of ischemic diseases because of their strong angiogenic actions. Although tissue ischemia is predominantly caused by atherosclerosis, the roles of endothelial FGF receptors (FGF-Rs) in atherosclerosis remain obscure. We generated endothelial cell (EC)-targeted constitutively active FGF-R2-overexpressing mice, using the Tie2 promoter (Tie2-FGF-R2-Tg), and crossed them with apolipoprotein E (ApoE)-deficient mice (ApoE-KO) to generate Tie2-FGF-R2-Tg/ApoE-deficient mice (Tie2-FGF-R2-Tg/ApoE-KO). After being fed a Western diet for 8 wk, the Tie2-FGF-R2-Tg/ApoE-KO demonstrated 2.0-fold greater atherosclerotic lesion area on the luminal surfaces of the aortas than the ApoE-KO (P < 0.01). The level of p21(Cip1) protein, a cell cycle inhibitor, in the FGF-R2-overexpressing EC was 2.5-fold greater than that in the wild-type (WT) EC at the baseline (P < 0.01). FGF-R2 overexpression in the EC resulted in increased expression of VCAM-1 and ICAM-1, acceleration of apoptosis, and decreased proliferative activity, all of which were normalized by small interfering RNA (siRNA)-mediated knockdown of p21(Cip1) (75% reduction in protein level, P < 0.01). Furthermore, the expression of PDGF-B and Egr-1, a PDGF/p21(Cip1)-inducible transcription factor, in the aortic endothelium of Tie2-FGF-R2-Tg/ApoE-KO was significantly greater than that in ApoE-KO. The proliferation of vascular smooth muscle cells in the aortic media of Tie2-FGF-R2-Tg/ApoE-KO was 2.0-fold higher than that in ApoE-KO (P < 0.01). Thus our study reveals that endothelial FGF-R2 signaling aggravates atherosclerosis by promoting p21(Cip1)-mediated EC dysfunction and cautions against the use of FGF for therapeutic angiogenesis in the setting of atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/physiology , Analysis of Variance , Animals , Aorta/metabolism , Aorta/physiopathology , Apoptosis , Atherosclerosis/physiopathology , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Diet , Endothelium, Vascular/physiopathology , Immunohistochemistry , Male , Mice , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...