Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Mod Rheumatol ; 33(1): 1-11, 2023 Jan 03.
Article in English | MEDLINE | ID: mdl-35535676

ABSTRACT

Pathological findings are important in the diagnosis of vasculitis. However, due to the rarity of the disease, standard textbooks usually devote only a few pages to this topic, and this makes it difficult for clinicians not specializing in vasculitis to fully understand the pathological findings in vasculitis. To address the paucity of information, we present representative pathological findings in vasculitis classified in the 2012 Revised International Chapel Hill Consensus Conference Nomenclature of Vasculitides (CHCC2012). The CHCC2012 classifies 26 vasculitides into seven categories: (1) large-vessel vasculitis, (2) medium-vessel vasculitis, (3) small-vessel vasculitis, including antineutrophil cytoplasmic antibody-associated vasculitis and immune complex small-vessel vasculitis, (4) variable-vessel vasculitis, (5) single-organ vasculitis, (6) vasculitis associated with systemic disease, and (7) vasculitis associated with probable aetiology. Moreover, representative pathological findings of vasculitis-related diseases and non-inflammatory vasculopathy not mentioned in the CHCC2012 are also presented. This will be useful for clinicians to refer to typical pathological findings of vasculitis in daily practice.


Subject(s)
Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Humans , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/complications , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/diagnosis , Consensus
2.
Nucleic Acids Res ; 45(15): 8758-8772, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28549158

ABSTRACT

Chromatin reorganization is necessary for pluripotent stem cells, including embryonic stem cells (ESCs), to acquire lineage potential. However, it remains unclear how ESCs maintain their characteristic chromatin state for appropriate gene expression upon differentiation. Here, we demonstrate that chromodomain helicase DNA-binding domain 2 (Chd2) is required to maintain the differentiation potential of mouse ESCs. Chd2-depleted ESCs showed suppressed expression of developmentally regulated genes upon differentiation and subsequent differentiation defects without affecting gene expression in the undifferentiated state. Furthermore, chromatin immunoprecipitation followed by sequencing revealed alterations in the nucleosome occupancy of the histone variant H3.3 for developmentally regulated genes in Chd2-depleted ESCs, which in turn led to elevated trimethylation of the histone H3 lysine 27. These results suggest that Chd2 is essential in preventing suppressive chromatin formation for developmentally regulated genes and determines subsequent effects on developmental processes in the undifferentiated state.


Subject(s)
Cell Differentiation/genetics , Chromatin Assembly and Disassembly/genetics , DNA-Binding Proteins/physiology , Mouse Embryonic Stem Cells/physiology , Animals , Cell Proliferation/genetics , Cells, Cultured , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Mice, Inbred NOD , Mice, SCID
3.
J Vasc Surg ; 68(2): 576-586.e1, 2018 08.
Article in English | MEDLINE | ID: mdl-28974341

ABSTRACT

OBJECTIVES: Budding uninhibited by benzimidazole-related 1 (BubR1), a cell cycle-related protein, is an essential component of the spindle checkpoint that regulates cell division. Mice in which BubR1 expression is reduced to 10% of the normal level display the phenotypic features of progeria. However, the role of BubR1 in vascular diseases and angiogenesis remains unknown. To investigate the influence of BubR1 on angiogenesis, we generated a low-null-BubR1-expressing (BubR1L/-) mouse strain with reduced BubR1 expression as low as 15% of the normal level without any abnormalities in appearance. METHODS: To elucidate the role of BubR1 in angiogenesis, we used a hind limb ischemia model induced in BubR1L/- mice and age-matched wild-type (WT) littermates. To evaluate the pathologic influence of BubR1 on angiogenesis, we measured the blood flow before and after hind limb ischemia surgery, and the expression of typical angiogenic factors in vivo and in vitro. RESULTS: In WT mice, blood flow in the ischemic left limb gradually recovered to approximately 80%, 14 days after surgery. Conversely, in the BubR1L/- group, blood flow in the left ischemic limb recovered to at most 30% (14 days after surgery, P < .01; immediately after the operation, and 5 and 9 days after surgery, P < .05). In adductor and calf muscles from BubR1L/- mice, regenerated muscle bundles, granulation tissue, and inflammatory cell invasion were more evident than in calf muscles from WT mice at 14 days after surgery. All WT mice at 14 days after surgery had complete limb salvage, but loss of limbs was observed in approximately 70% of BubR1L/- mice (P < .05). The vascular endothelial growth factor protein increase in ischemic hind limb muscles was lower in BubR1L/- mice compared with WT mice (P < .05), and vascular endothelial growth factor levels in human aortic smooth muscle cells treated with BubR1 knockdown siRNA were lower compared with scramble siRNA under hypoxic conditions (P < .01). HIF1α protein levels in the muscles after hind limb ischemia surgery were also significantly lower in BubR1L/- mice compared with WT mice (P < .05). CONCLUSIONS: BubR1 insufficiency impairs angiogenesis and results in limb loss in ischemic hind limbs. BubR1 may be a crucial angiogenic factor and might be beneficial for the treatment of limb ischemia.


Subject(s)
Aging/metabolism , Cell Cycle Proteins/deficiency , Ischemia/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Protein Serine-Threonine Kinases/deficiency , Age Factors , Aging/genetics , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Critical Illness , Disease Models, Animal , Hindlimb , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Ischemia/genetics , Ischemia/physiopathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Regional Blood Flow , Signal Transduction , Time Factors , Transfection , Vascular Endothelial Growth Factor A/metabolism
4.
J Immunol ; 196(6): 2779-87, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26880761

ABSTRACT

Intrauterine fetal growth restriction (IUGR) and death (IUFD) are both serious problems in the perinatal medicine. Fetal vasculopathy is currently considered to account for a pathogenic mechanism of IUGR and IUFD. We previously demonstrated that an innate immune receptor, the nucleotide-binding oligomerization domain-1 (Nod1), contributed to the development of vascular inflammations in mice at postnatal stages. However, little is known about the deleterious effects of activated Nod1 signaling on embryonic growth and development. We report that administration of FK565, one of the Nod1 ligands, to pregnant C57BL/6 mice induced IUGR and IUFD. Mass spectrometry analysis revealed that maternally injected FK565 was distributed to the fetal tissues across placenta. In addition, maternal injection of FK565 induced robust increases in the amounts of CCL2, IL-6, and TNF proteins as well as NO in maternal, placental and fetal tissues. Nod1 was highly expressed in fetal vascular tissues, where significantly higher levels of CCL2 and IL-6 mRNAs were induced with maternal injection of FK565 than those in other tissues. Using Nod1-knockout mice, we verified that both maternal and fetal tissues were involved in the development of IUGR and IUFD. Furthermore, FK565 induced upregulation of genes associated with immune response, inflammation, and apoptosis in fetal vascular tissues. Our data thus provided new evidence for the pathogenic role of Nod1 in the development of IUGR and IUFD at the maternal-fetal interface.


Subject(s)
Fetal Death/prevention & control , Fetal Growth Retardation/immunology , Nod1 Signaling Adaptor Protein/metabolism , Oligopeptides/administration & dosage , Vasculitis/immunology , Animals , Chemokine CCL2/metabolism , Female , Fetal Death/etiology , Fetal Growth Retardation/chemically induced , Humans , Interleukin-6/metabolism , Ligands , Maternal Exposure/adverse effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/genetics , Pregnancy , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Vasculitis/chemically induced
5.
J Immunol ; 194(2): 773-80, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25488987

ABSTRACT

Atherosclerosis is essentially a vascular inflammatory process in the presence of an excess amount of lipid. We have recently reported that oral administration of a nucleotide-binding oligomerization domain (Nod)-1 ligand, FK565, induced vascular inflammation in vivo. No studies, however, have proven the association between Nod1 and atherosclerosis in vivo. To investigate a potential role of NOD1 in atherogenesis, we orally administered FK565 to apolipoprotein E knockout (Apoe(-/-)) mice for 4 wk intermittently and performed quantification of atherosclerotic lesions in aortic roots and aortas, immunohistochemical analyses, and microarray-based gene expression profiling of aortic roots. FK565 administration accelerated the development of atherosclerosis in Apoe(-/-) mice, and the effect was dependent on Nod1 in non-bone marrow origin cells by bone marrow transplantation experiments. Immunohistochemical studies revealed the increases in the accumulation of macrophages and CD3 T cells within the plaques in aortic roots. Gene expression analyses of aortic roots demonstrated a marked upregulation of the Ccl5 gene during early stage of atherogenesis, and the treatment with Ccl5 antagonist significantly inhibited the acceleration of atherosclerosis in FK565-administered Apoe(-/-) mice. Additionally, as compared with Apoe(-/-) mice, Apoe and Nod1 double-knockout mice showed reduced development of atherosclerotic lesions from the early stage as well as their delayed progression and a significant reduction in Ccl5 mRNA levels at 9 wk of age. Data in the present study show that the Nod1 signaling pathway in non-bone marrow-derived cells contributes to the development of atherosclerosis.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/immunology , Bone Marrow Cells/immunology , Macrophages/immunology , Nod1 Signaling Adaptor Protein/immunology , T-Lymphocytes/immunology , Adjuvants, Immunologic/pharmacology , Animals , Aorta/immunology , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/pathology , Macrophages/pathology , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Oligopeptides/pharmacology , T-Lymphocytes/pathology
6.
Arterioscler Thromb Vasc Biol ; 35(2): 341-7, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25524773

ABSTRACT

OBJECTIVE: BubR1, a cell cycle-related protein, is an essential component of the spindle checkpoint that regulates cell division. Mice with BubR1 expression reduced to 10% of the normal level display a phenotype characterized by progeria; however, the involvement of BubR1 in vascular diseases is still unknown. We generated mice in which BubR1 expression was reduced to 20% (BubR1(L/L) mice) of that in wild-type mice (BubR1(+/+)) to investigate the effects of BubR1 on arterial intimal hyperplasia. APPROACH AND RESULTS: Ten-week-old male BubR1(L/L) and age-matched wild-type littermates (BubR1(+/+)) were used in this study. The left common carotid artery was ligated, and histopathologic examinations were conducted 4 weeks later. Bone marrow transplantation was also performed. Vascular smooth muscle cells (VSMCs) were isolated from the thoracic aorta to examine cell proliferation, migration, and cell cycle progression. Severe neointimal hyperplasia was observed after artery ligation in BubR1(+/+) mice, whereas BubR1(L/L) mice displayed nearly complete inhibition of neointimal hyperplasia. Bone marrow transplantation from all donors did not affect the reconstitution of 3 hematopoietic lineages, and neointimal hyperplasia was still suppressed after bone marrow transplantation from BubR1(+/+) mice to BubR1(L/L) mice. VSMC proliferation was impaired in BubR1(L/L) mice because of delayed entry into the S phase. VSMC migration was unaffected in these BubR1(L/L) mice. p38 mitogen-activated protein kinase-inhibited VSMCs showed low expression of BubR1, and BubR1-inhibited VSMCs showed low expression of p38. CONCLUSIONS: BubR1 may represent a new target molecule for treating pathological states of vascular remodeling, such as restenosis after angioplasty.


Subject(s)
Carotid Artery Diseases/metabolism , Cell Cycle Proteins/deficiency , Cell Proliferation , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima , Protein Serine-Threonine Kinases/deficiency , Animals , Bone Marrow Transplantation , Carotid Artery Diseases/genetics , Carotid Artery Diseases/pathology , Carotid Artery, Common/metabolism , Carotid Artery, Common/pathology , Carotid Artery, Common/surgery , Cell Cycle Proteins/genetics , Cell Movement , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Hyperplasia , Ligation , Male , Mice, 129 Strain , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/surgery , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , RNA Interference , S Phase Cell Cycle Checkpoints , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Mol Ther ; 21(3): 707-14, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23319060

ABSTRACT

We here report the results of a Phase I/IIa open-label four dose-escalation clinical study assessing the safety, tolerability, and possible therapeutic efficacy of a single intramuscular administration of DVC1-0101, a new gene transfer vector based on a nontransmissible recombinant Sendai virus (rSeV) expressing the human fibroblast growth factor-2 (FGF-2) gene (rSeV/dF-hFGF2), in patients with peripheral arterial disease (PAD). Gene transfer was done in 12 limbs of 12 patients with rest pain, and three of them had ischemic ulcer(s). No cardiovascular or other serious adverse events (SAEs) caused by gene transfer were detected in the patients over a 6-month follow-up. No infectious viral particles, as assessed by hemagglutination activity, were detected in any patient during the study. No representative elevation of proinflammatory cytokines or plasma FGF-2 was seen. Significant and continuous improvements in Rutherford category, absolute claudication distance (ACD), and rest pain were observed (P < 0.05 to 0.01). To the best of our knowledge, this is the first clinical trial of the use of a gene transfer vector based on rSeV. The single intramuscular administration of DVC1-0101 to PAD patients was safe and well tolerated, and resulted in significant improvements of limb function. Larger pivotal studies are warranted as a next step.


Subject(s)
Fibroblast Growth Factor 2/genetics , Genetic Therapy/methods , Peripheral Arterial Disease/therapy , Aged , Aged, 80 and over , Cytokines/metabolism , Female , Gene Transfer Techniques , Genetic Vectors , Humans , Injections, Intramuscular , Male , Middle Aged , Peripheral Arterial Disease/genetics , Sendai virus/genetics , Treatment Outcome
9.
Kidney Int ; 84(2): 373-80, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23594677

ABSTRACT

There is little information regarding whether patients with chronic kidney disease (CKD) have a high incidence of vulnerable plaques in their coronary arteries. To gain additional evidence on this, we conducted a population-based study by randomly selecting 126 subjects from 844 consecutive autopsies of elderly residents of Hisayama, Japan. We then determined the relationships of CKD with neovascularization and intraplaque hemorrhage in coronary atherosclerosis with the subjects classified into four categories based on their estimated glomerular filtration rate (eGFR). Areas of oxidized low-density lipoprotein (oxLDL) and vascular endothelial growth factor (VEGF) expression, assessed by immunohistochemistry in a total of 375 coronary arteries, increased significantly with decreasing eGFR. A lower eGFR was also associated with increased numbers of newly formed blood vessels. These relationships remained substantially unchanged after adjustment for confounding factors. The multivariate-adjusted odds ratio of the presence of intraplaque hemorrhages was 6.2 (95% confidence interval, 1.1-35.0) in patients with an eGFR <30 ml/min/1.73 m(2) compared with those with an eGFR of ≥ 60 ml/min/1.73 m(2). Thus, elderly patients with CKD have intimal neoangiogenesis and an increased risk of intraplaque hemorrhage in coronary arteries, possibly favored by local accumulation of oxLDL and VEGF.


Subject(s)
Coronary Artery Disease/epidemiology , Coronary Vessels/pathology , Hemorrhage/epidemiology , Neovascularization, Pathologic , Plaque, Atherosclerotic , Renal Insufficiency, Chronic/epidemiology , Adult , Age Factors , Aged , Aged, 80 and over , Autopsy , Biomarkers/analysis , Coronary Artery Disease/metabolism , Coronary Artery Disease/mortality , Coronary Artery Disease/pathology , Coronary Vessels/chemistry , Cross-Sectional Studies , Female , Glomerular Filtration Rate , Hemorrhage/metabolism , Hemorrhage/mortality , Hemorrhage/pathology , Humans , Immunohistochemistry , Incidence , Japan/epidemiology , Kidney/physiopathology , Linear Models , Lipoproteins, LDL/analysis , Logistic Models , Male , Middle Aged , Multivariate Analysis , Odds Ratio , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/mortality , Renal Insufficiency, Chronic/physiopathology , Risk Factors , Rupture, Spontaneous , Severity of Illness Index , Vascular Endothelial Growth Factor A/analysis , Young Adult
10.
Mol Ther ; 20(4): 769-77, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22314292

ABSTRACT

Malignant pleural mesothelioma (MPM) is highly intractable and readily spreads throughout the surface of the pleural cavity, and these cells have been shown to express urokinase-type plasminogen activator receptor (uPAR). We here examined the potential of our new and powerful recombinant Sendai virus (rSeV), which shows uPAR-specific cell-to-cell fusion activity (rSeV/dMFct14 (uPA2), named "BioKnife"), for tumor cell killing in two independent orthotopic xenograft models of human. Multicycle treatment using BioKnife resulted in the efficient rescue of these models, in association with tumor-specific fusion and apoptosis. Such an effect was also seen on both MSTO-211H and H226 cells in vitro; however, we confirmed that the latter expressed uPAR but not uPA. Of interest, infection with BioKnife strongly facilitated the uPA release from H226 cells, and this effect was completely abolished by use of either pyrrolidine dithiocarbamate (PDTC) or BioKnife expressing the C-terminus-deleted dominant negative inhibitor for retinoic acid-inducible gene-I (RIG-IC), indicating that BioKnife-dependent expression of uPA was mediated by the RIG-I/nuclear factor-κB (NF-κB) axis, detecting RNA viral genome replication. Therefore, these results suggest a proof of concept that the tumor cell-killing mechanism via BioKnife may have significant potential to treat patients with MPM that is characterized by frequent uPAR expression in a clinical setting.


Subject(s)
Mesothelioma/metabolism , Mesothelioma/therapy , Oncolytic Viruses/physiology , Pleural Neoplasms/metabolism , Pleural Neoplasms/therapy , Sendai virus/physiology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Mesothelioma/genetics , Mice , Oncolytic Viruses/genetics , Pleural Neoplasms/genetics , RNA, Small Interfering , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus/genetics , Urokinase-Type Plasminogen Activator/genetics , Xenograft Model Antitumor Assays
11.
Mod Rheumatol Case Rep ; 8(1): 125-132, 2023 Dec 29.
Article in English | MEDLINE | ID: mdl-37319013

ABSTRACT

In response to the coronavirus disease 2019 pandemic, the coronavirus disease 2019 vaccine was rapidly developed and the effectiveness of the vaccine has been established. However, various adverse effects have been reported, including the development of autoimmune diseases. We report a case of new-onset polyarteritis nodosa in a 32-year-old male following the coronavirus disease 2019 vaccination. The patient developed limb pain, fever, pulmonary embolism, multiple subcutaneous nodules, and haematomas. Skin biopsy revealed necrotising inflammation accompanied by fibrinoid necrosis and high inflammatory cell infiltration in the walls of medium to small arteries. The symptoms resolved following corticosteroid treatment. Although it is difficult to prove a relationship between the vaccine and polyarteritis nodosa, similar cases have been reported and further reports and analyses are therefore necessary.


Subject(s)
COVID-19 Vaccines , COVID-19 , Polyarteritis Nodosa , Adult , Humans , Male , Adrenal Cortex Hormones , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19/complications , COVID-19 Vaccines/adverse effects , Polyarteritis Nodosa/diagnosis , Polyarteritis Nodosa/etiology , Skin/pathology
12.
Neurourol Urodyn ; 31(4): 593-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22396250

ABSTRACT

AIMS: We investigated the relative important role of rho kinase (ROK) and protein kinase C (PKC) pathways in carbachol (CCh)-induced Ca(2+) sensitization in α-toxin permeabilized Guinea pig detrusor smooth muscle (DSM) following bladder outlet obstruction (BOO). METHODS: Bladder outlet obstruction was created by placement of a silver jeweler's jump rings loosely round the urethro-vesical junction of Guinea pigs. Sham operated Guinea pig underwent a similar protocol without application of the ring and served as control. α-Toxin permeabilized DSM strips from control Guinea pigs and those subjected to 6-8 weeks of BOO were mounted horizontally for isometric force recording in 100 µl relaxing solution on perspex block. The effect of ROK inhibitor (Y-27632) and PKC inhibitor (GF-109203X) on CCh-induced Ca(2+) sensitization was studied during sustained contraction. Permeabilized DSM strips were also stimulated by cumulative increase of Ca(2+) concentration compared to that in control in the presence and in the absence of sensitization-induced PKC activator, phorbol 12,13-dibutyrate. RESULTS: Ca(2+) sensitization-induced by CCh was greater in BOO compared to controls. This muscarinic agonist-induced Ca(2+) sensitization was inhibited by Y-27632 or GF-109203X. The inhibitory effect of Y-27632 (5 µM) was greater while the inhibitory effect of GF-109203X (5 µM) was smaller in BOO compared to that in controls. Phorbol 12,13-dibutyrate (1 µM) markedly increased Ca(2+) sensitivity in controls but not in BOO. CONCLUSIONS: Our findings provide the first evidence that BOO enhances the ROK pathway and diminishes the PKC pathway in CCh-induced Ca(2+) sensitization in contraction of permeabilized Guinea pig DSM and suggest that inhibitors of ROK might potentially relieve bladder dysfunction related to BOO.


Subject(s)
Calcium/metabolism , Muscle Contraction/physiology , Muscle, Smooth/physiology , Protein Kinase C/metabolism , Signal Transduction/physiology , Urinary Bladder Neck Obstruction/metabolism , rho-Associated Kinases/metabolism , Amides/pharmacology , Animals , Carbachol/pharmacology , Enzyme Inhibitors/pharmacology , Guinea Pigs , Indoles/pharmacology , Male , Maleimides/pharmacology , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Protein Kinase C/antagonists & inhibitors , Pyridines/pharmacology , Signal Transduction/drug effects , rho-Associated Kinases/antagonists & inhibitors
13.
J Oral Pathol Med ; 41(9): 675-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22582814

ABSTRACT

Vasculature development is thought to be an important aspect in the growth and metastasis of solid tumors. Among the many angiogenic factors produced by tumor cells, vascular endothelial growth factor (VEGF) is considered to play a key role in angiogenic processes. VEGF synthesis is modulated by hypoxia-inducible factor-1 (HIF-1) function within the hypoxic microenvironment of growing cancer tissue. To inhibit HIF-1 activation, oligodeoxynucleotides (ODNs) were synthesized and transferred with either the consensus sequence for HIF-1 binding or a mutated form of this sequence. If we could transfer a large number of ODNs into the cancer cell nucleus, activated HIF-1 might bind to the ODNs, resulting in inhibition of hypoxia-induced VEGF synthesis. We transferred these ODNs into cultured oral squamous cell carcinoma cells (SAS cells) using the hemagglutinating virus of Japan (HVJ)-liposome method. Hypoxia-mediated expression of VEGF by cancer cells was suppressed by transfection of HIF-1 decoy ODNs, but not by mutated HIF-1 decoy ODNs. HIF-1 decoy ODN transfection also inhibited VEGF protein synthesis. These results suggest that transfection with HIF-1 decoy ODNs is effective for regulating tumor growth by reducing VEGF.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Hypoxia-Inducible Factor 1/genetics , Mouth Neoplasms/metabolism , Oligodeoxyribonucleotides/genetics , Transfection/methods , Vascular Endothelial Growth Factor A/metabolism , Carcinoma, Squamous Cell/genetics , Cell Culture Techniques , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Nucleus/metabolism , Consensus Sequence/genetics , Cytoplasm/metabolism , Genetic Vectors/genetics , Humans , Liposomes , Mouth Neoplasms/genetics , Mutation/genetics , Neovascularization, Pathologic/genetics , Promoter Regions, Genetic/genetics , Sendai virus/genetics , Transcriptional Activation/genetics , Vascular Endothelial Growth Factor A/analysis
14.
Arterioscler Thromb Vasc Biol ; 30(4): 818-26, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20056911

ABSTRACT

OBJECTIVE: The angiopoietin (Ang)-Tie-2 system plays a critical role during fetal and adult angiogenesis. Herein, we explored the Tie-2 shedding-related molecular mechanisms and the pathophysiological significance. METHODS AND RESULTS: By using a mouse hindlimb ischemia model, we observed dissociated expression between the full-length Tie-2 (fTie-2) protein and Tie-2 mRNA in thigh muscles 1 day after an ischemic operation, suggesting that fTie-2 expression was modified through the posttranscriptional regulation in vivo. A soluble form of Tie-2 produced in human umbilical vein endothelial cells was dramatically suppressed by treatment with siRNA-matrix metalloproteinase (MMP) 14 or tissue inhibitor of metalloproteinase 3, resulting in an increase in cellular fTie-2 and thereby enhancing Ang-1-dependent Akt phosphorylation and Akt-dependent endothelial functions, such as Ang-2 downregulation or an increase of endothelial viability. Phorbol-12-myristate-13 acetate (PMA) upregulates MMP-14 mRNA via protein kinase C-extracellular signal-regulated kinase pathways, and enhanced soluble Tie-2 production in an MMP-14-dependent manner, resulting in a reduction of cellular fTie-2. In addition, the PMA-induced soluble Tie-2 was mediated by the protein kinase C-extracellular signal-regulated kinase signaling pathways. Finally, downregulation of tissue inhibitor of metalloproteinase 3 and upregulation of MMP-14 mRNA were confirmed in ischemic thigh muscles 1 day after the operation. CONCLUSIONS: An autocrine linkage between the endothelial protein kinase C-MMP-14 axis and Tie-2 shedding was shown to be a novel regulatory mechanism for the Ang-Tie-2 system and may play a role in modulating endothelial function during angiogenesis.


Subject(s)
Angiopoietin-1/metabolism , Autocrine Communication , Endothelial Cells/enzymology , Ischemia/enzymology , Matrix Metalloproteinase 14/metabolism , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Receptor, TIE-2/metabolism , Animals , Autocrine Communication/drug effects , Cells, Cultured , Disease Models, Animal , Endothelial Cells/drug effects , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Hindlimb , Humans , Ischemia/physiopathology , Male , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Protease Inhibitors/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA Processing, Post-Transcriptional , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, TIE-2/genetics , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Tissue Inhibitor of Metalloproteinases/metabolism , Transfection
15.
Mol Ther ; 18(10): 1778-86, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20606645

ABSTRACT

Glioblastoma multiforme (GM), the most frequent primary malignant brain tumor, is highly invasive due to the expression of proteases, including urokinase-type plasminogen activator (uPA). Here, we show the potential of our new and powerful recombinant Sendai virus (rSeV) showing uPA-specific cell-to-cell fusion activity [rSeV/dMFct14 (uPA2), named "BioKnife"] for GM treatment, an effect that was synergistically enhanced by arming BioKnife with the interferon-ß (IFN-ß) gene. BioKnife killed human GM cell lines efficiently in a uPA-dependent fashion, and this killing was prevented by PA inhibitor-1. Rat gliosarcoma 9L cells expressing both uPA and its functional receptor uPAR (9L-L/R) exhibited high uPA activity on the cellular surface and were highly susceptible to BioKnife. Although parent 9L cells (9L-P) were resistant to BioKnife and to BioKnife expressing IFN-ß (BioKnife-IFNß), cell-cell fusion of 9L-L/R strongly facilitated the expression of IFN-ß, and in turn, IFN-ß significantly accelerated the fusion activity of BioKnife. A similar synergy was seen in a rat orthotopic brain GM model with 9L-L/R in vivo; therefore, these results suggest that BioKnife-IFNß may have significant potential to improve the survival of GM patients in a clinical setting.


Subject(s)
Glioblastoma/therapy , Interferon-beta/metabolism , Oncolytic Viruses/physiology , Sendai virus/physiology , Urokinase-Type Plasminogen Activator/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Enzyme-Linked Immunosorbent Assay , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Interferon-beta/genetics , Magnetic Resonance Imaging , Mice , Mice, Nude , Oncolytic Viruses/genetics , Plasminogen Activator Inhibitor 1/pharmacology , Rats , Rats, Inbred F344 , Receptors, Urokinase Plasminogen Activator/genetics , Receptors, Urokinase Plasminogen Activator/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus/genetics , Urokinase-Type Plasminogen Activator/genetics
16.
Mol Cancer ; 9: 287, 2010 Oct 31.
Article in English | MEDLINE | ID: mdl-21034514

ABSTRACT

BACKGROUND: Podoplanin, a mucin-like transmembrane glycoprotein, is reportedly expressed in a variety of malignant cells and is generally regarded as a factor for promoting tumor progression in conventional studies. By contrast, a clinicopathologically conflicting role for podoplanin, namely as a favorable prognostic factor for patients with lung/cervical squamous cell carcinoma (SCC), has recently been reported. Here, we investigated the role of podoplanin expressed in lung squamoid cancer cells (LSCCs) in experimental tumor progression. RESULTS: Using EBC-1 cells, a lung SCC cell line without podoplanin expression and with lymphogenous metastatic potential, stable transformants with or without an exogenous human podoplanin gene were established and applied to a mouse tumor implantation model. In vivo examinations revealed that exogenous podoplanin had no influence on tumor growth, whereas it significantly restrained axillary lymph node metastasis associated with the suppression of lymphangiogenesis but not angiogenesis and with the downregulation of EBC-1-derived VEGF-C but not other lymphangiogenesis-related factor mRNAs in implanted tumor tissue. In vitro examinations to clarify the mechanisms underlying the in vivo phenomena revealed that exogenous podoplanin significantly suppressed the expression of VEGF-C mRNA and of the protein, and also increased the level of phosphorylated c-jun N terminal kinase (JNK) in EBC-1 cells. The former effect of exogenous podoplanin was impaired by treatment with either JNK inhibitor sp600125 or podoplanin-siRNA, and the latter effect was impaired by treatment with podoplanin-siRNA, suggesting that podoplanin was able to activate JNK, thereby downregulating VEGF-C gene expression in LSCCs (podoplanin-JNK-VEGF-C axis). Furthermore, supporting evidence in regard to the axis present in LSCCs was obtained from similar experiments using H157 cells, another lung SCC cell line expressing endogenous podoplanin. CONCLUSIONS: Our findings suggested that LSCC-associated podoplanin was functional and could attenuate the potential for lymph node metastasis, possibly based on the suppression of tumor lymphangiogenesis; thus, podoplanin in cancer cells may become a useful biomarker to measure the malignancy of lung SCC.


Subject(s)
Lung Neoplasms/metabolism , Membrane Glycoproteins/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Electrophoretic Mobility Shift Assay , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Male , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
17.
Am J Pathol ; 175(4): 1709-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19779139

ABSTRACT

Numerous past studies have suggested a critical role of the paracrine effect between tumor vascular endothelial growth factor (VEGF)-C and lymphatic FLT-4 in solid tumor-associated lymphangiogenesis. In contrast, the pathophysiological role of tumor cell-associated FLT-4 in tumor progression remains to be elucidated. Here, we investigated this role using a tumor implantation model. SAS cells, an oral squamous carcinoma cell line expressing both VEGF-C and FLT-4 but neither FLK-1/KDR nor VEGF-D were adopted for experiments. Stable transformants of dominant-negative (dn) SAS cells were established in which the cytoplasmic domain-deleted FLT-4 was exogenously overexpressed, which can lead to inactivation of endogenous FLT-4 through competitive antagonism and is associated with down-activation of endogenous FLT-4-related intracellular signals. In vitro and in vivo proliferation assays showed lower proliferative activity of dn-SAS cells. An immunohistochemical study revealed that the tumor lymphangiogenesis was significantly suppressed, and the level of human VEGF-C mRNA was significantly lower in dn-SAS cell-derived tumor tissues. Moreover, in vitro studies demonstrated that the significant suppression of VEGF-C and VEGF-A expression was evident in dn-SAS cells or wild-type SAS cells treated with either the FLT-4 kinase inhibitor MAZ51 or the inhibitor of FLT-4-related signals. These findings together suggested that the VEGF-C/FLT-4 autocrine loop in tumor cells was a potential enhancer system to promote cancer progression, and FLT-4 in tumor tissue might become an effective target for cancer therapy.


Subject(s)
Autocrine Communication , Carcinoma, Squamous Cell/enzymology , Lymphangiogenesis , Mouth Neoplasms/enzymology , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Animals , Autocrine Communication/drug effects , Carcinoma, Squamous Cell/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Lymphangiogenesis/drug effects , Male , Mice , Mouth Neoplasms/genetics , Naphthalenes/pharmacology , Neoplasm Transplantation , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors
18.
Am J Physiol Heart Circ Physiol ; 297(5): H1685-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19734356

ABSTRACT

Emerging evidence indicates that the tight communication between vascular endothelial cells and mural cells using platelet-derived growth factor (PDGF)-BB is essential for capillary stabilization during the angiogenic process. However, little is known about the related regulator that determines PDGF-BB expression. Using murine models of therapeutic neovascularization, we here show that a typical lymphangiogenic factor, vascular endothelial growth factor (VEGF)-C, is an essential regulator determining PDGF-BB expression for vascular stabilization via a paracrine mode of action. The blockade of VEGF type 3 receptor (VEGFR3) using neutralizing antibody AFL-4 abrogated FGF-2-mediated limb salvage and blood flow recovery in severely ischemic hindlimb. Interestingly, inhibition of VEGFR3 activity not only diminished lymphangiogenesis, but induced marked dilatation of capillary vessels, showing mural cell dissociation. In these mice, VEGF-C and PDGF-B were upregulated in the later phase after induced ischemia, on day 7, when exogenous FGF-2 expression had already declined, and blockade of VEGFR3 or PDGF-BB activities diminished PDGF-B or VEGF-C expression, respectively. These results clearly indicate that VEGF-C is a critical mediator, not only for lymphangiogenesis, but also for capillary stabilization, the essential molecular mechanism of communication between endothelial cells and mural cells during neovascularization.


Subject(s)
Capillaries/metabolism , Ischemia/metabolism , Lymphangiogenesis , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Proto-Oncogene Proteins c-sis/metabolism , Vascular Endothelial Growth Factor C/metabolism , Amputation, Surgical , Animals , Antibodies/pharmacology , Becaplermin , Capillaries/drug effects , Capillaries/physiopathology , Cattle , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Feedback, Physiological , Fibroblast Growth Factor 2/biosynthesis , Fibroblast Growth Factor 2/genetics , Genetic Therapy , Hindlimb , Humans , Ischemia/genetics , Ischemia/physiopathology , Ischemia/therapy , Lymphangiogenesis/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neovascularization, Physiologic/drug effects , Paracrine Communication , Platelet-Derived Growth Factor/metabolism , Receptor, Platelet-Derived Growth Factor beta/metabolism , Regional Blood Flow , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/metabolism
19.
Am J Pathol ; 173(5): 1326-38, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18845835

ABSTRACT

Photoreceptor apoptosis is a critical process of retinal degeneration in retinitis pigmentosa (RP), a group of retinal degenerative diseases that result from rod and cone photoreceptor cell death and represent a major cause of adult blindness. We previously demonstrated the efficient prevention of photoreceptor apoptosis by intraocular gene transfer of pigment epithelium-derived factor (PEDF) in animal models of RP; however, the underlying mechanism of the neuroprotective activity of PEDF remains elusive. In this study, we show that an apoptosis-inducing factor (AIF)-related pathway is an essential target of PEDF-mediated neuroprotection. PEDF rescued serum starvation-induced apoptosis, which is mediated by AIF but not by caspases, of R28 cells derived from the rat retina by preventing translocation of AIF into the nucleus. Nuclear translocation of AIF was also observed in the apoptotic photoreceptors of Royal College of Surgeons rats, a well-known animal model of RP that carries a mutation of the Mertk gene. Lentivirus-mediated retinal gene transfer of PEDF prevented the nuclear translocation of AIF in vivo, resulting in the inhibition of the apoptotic loss of their photoreceptors in association with up-regulated Bcl-2 expression, which mediates the mitochondrial release of AIF. These findings clearly demonstrate that AIF is an essential executioner of photoreceptor apoptosis in inherited retinal degeneration and provide a therapeutic rationale for PEDF-mediated neuroprotective gene therapy for individuals with RP.


Subject(s)
Apoptosis Inducing Factor/metabolism , Cell Nucleus/metabolism , Cytoprotection , Eye Proteins/metabolism , Nerve Growth Factors/metabolism , Neurons/cytology , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Serpins/metabolism , Animals , Apoptosis/drug effects , Apoptosis Inducing Factor/genetics , Caspases/metabolism , Cell Line , Cell Nucleus/drug effects , Cytoprotection/drug effects , Disease Models, Animal , Eye Proteins/pharmacology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Nerve Growth Factors/pharmacology , Neurons/drug effects , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Serpins/pharmacology , Serum , Signal Transduction/drug effects , Transduction, Genetic , Up-Regulation/drug effects
20.
Circ Res ; 98(1): 55-62, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16306442

ABSTRACT

Diabetic foot is caused by microangiopathy and is suggested to be a result of impaired angiogenesis. Using a severe hindlimb ischemia model of streptozotocin-induced diabetic mice (STZ-DM), we show that diabetic foot is a disease solely of the disturbance of platelet-derived growth factor B-chain homodimer (PDGF-BB) expression but not responses of angiogenic factors. STZ-DM mice frequently lost their hindlimbs after induced ischemia, whereas non-DM mice did not. Screening of angiogenesis-related factors revealed that only the expression of PDGF-BB was impaired in the STZ-DM mice on baseline, as well as over a time course after limb ischemia. Supplementation of the PDGF-B gene resulted in the prevention of autoamputation, and, furthermore, a protein kinase C (PKC) inhibitor restored the PDGF-BB expression and also resulted in complete rescue of the limbs of the STZ-DM mice. Inhibition of overproduction of advanced-glycation end product resulted in dephosphorylation of PKC-alpha and restored expression of PDGF-BB irrespective of blood sugar and HbA1c, indicating that advanced-glycation end product is an essential regulator for PKC/PDGF-BB in diabetic state. These findings are clear evidence indicating that diabetic vascular complications are caused by impairment of the PKC/PDGF-B axis, but not by the impaired expression of angiogenic factors, and possibly imply the molecular target of diabetic foot.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/etiology , Hindlimb/blood supply , Ischemia/physiopathology , Platelet-Derived Growth Factor/physiology , Protein Kinase C/physiology , Animals , Becaplermin , Diabetic Angiopathies/physiopathology , Fibroblast Growth Factor 2/physiology , Glycation End Products, Advanced/physiology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic , Phosphorylation , Proto-Oncogene Proteins c-sis , Streptozocin
SELECTION OF CITATIONS
SEARCH DETAIL