Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Life Sci ; 288: 120171, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34822800

ABSTRACT

AIM: The abnormal expression of oncogenic tyrosine kinase receptors such as platelet-derived growth factor receptors (PDGFRs) has been reported in cancer progression. However, the role of PDGFRs in the human androgen-independent prostate cancer PC-3 cell line is not well understood. Thus, this study examined the role of PDGFRs in androgen-independent PC-3 cells. MAIN METHODS: PDGFR mRNA and protein expression was determined by quantitative real-time PCR and western blotting, respectively. The effects of the tyrosine kinase inhibitor imatinib (imatinib mesylate) and small interfering RNAs (siRNAs) were determined by a Cell Counting Kit-8 assay, bromodeoxyuridine assay, and Transwell migration assay. The in vivo effect of imatinib was analyzed using a tumor formation assay in nude mice. KEY FINDINGS: PDGFRα was upregulated in androgen-independent PC-3 cells compared with normal prostate epithelial cells. PDGF-BB induced the phosphorylation of PDGFRα and downstream signaling molecules, including Akt, in a dose-dependent manner. Imatinib reduced the phosphorylation of the PDGFRα/Akt axis. Imatinib also suppressed the viability, proliferation, migration, and tumor growth of PC-3 cells. PDGFRα knockdown by siRNA decreased the viability and migration of PC-3 cells. SIGNIFICANCE: These results demonstrated the distinct contribution of PDGFRα signaling to the proliferation and migration of PC-3 cells and suggested the potential for PDGFRα as a therapeutic target for metastatic and androgen-independent prostate cancer.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Imatinib Mesylate/pharmacology , Prostatic Neoplasms/prevention & control , RNA, Small Interfering/genetics , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Int J Biometeorol ; 65(7): 1119-1124, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33580836

ABSTRACT

Various biological processes show seasonal variations in humans, including important metabolic pathways. Seasonal changes in gene expression may affect annual differences in human immunity. We hypothesized that seasonal changes occur in clock gene expression levels that are associated with body mass index. Six non-obese men and five obese men participated in summer and winter. Two milliliters of saliva were collected, and total RNA was isolated from buccal epithelial cells in saliva. The clock gene expression levels of CLOCK, BMAL1, PER1, CRY2, REV-ERB-α, and REV-ERB-ß were examined by real-time PCR. Blood samples were measured HbA1c, glucose, insulin, adiponectin, IL-6, and TNF-α. Participants were asked about their sleeping hours and seasonal pattern. In the present study, CLOCK, BMAL1, and REV-ERB-ß gene expression levels were significantly lower in winter than in summer; BMAL1 expression level was significantly lower in obese men than in non-obese. Concentrations of adiponectin and insulin were significantly different between obese and non-obese. No significant seasonal effects were observed in HbA1c, glucose, insulin, adiponectin, IL-6, or TNF-α concentrations. Sleep duration did not significantly differ between summer and winter. The short photoperiod during winter might contribute to seasonal alterations in the expression of clock genes in men. In the present results, revealed seasonal differences in clock gene expression levels might be associated with obesity. These results also showed the potential for measuring clock gene expression in a non-invasive manner using saliva samples.


Subject(s)
Circadian Rhythm , Obesity , CLOCK Proteins/genetics , Circadian Rhythm/genetics , Epithelial Cells , Gene Expression , Humans , Male , Obesity/genetics , Pilot Projects , Seasons
3.
J Physiol Sci ; 70(1): 44, 2020 Sep 22.
Article in English | MEDLINE | ID: mdl-32962633

ABSTRACT

Glucose uptake and adenosine triphosphate (ATP) generation are important for the survival and growth of endothelial cells. An increase of glucose uptake under hypoxia was previously shown to be associated with the increased expression of glucose transporters (GLUTs). However, the regulation of GLUT trafficking to the cell surface has not been examined in detail. Here, we report the characterization of GLUT1 translocation to the plasma membrane during hypoxia in endothelial cells. Human umbilical vein endothelial cells (HUVECs) were exposed to hypoxia (1% O2) for 12 h, which significantly induced GLUT1 expression and translocation to the plasma membrane. GLUT1 translocation was associated with a decrease of intracellular ATP by hypoxia. Decreasing ATP levels with antimycin-A and 2-deoxyglucose induced GLUT1 translocation under normoxia. The induction of hypoxia-inducible factor-1α under normoxia did not influence the cell surface expression of GLUT1 or cellular ATP concentration. Interestingly, the translocation of GLUT1 induced by hypoxia was inhibited by the ATP-sensitive potassium (KATP) channel inhibitor glibenclamide, while the mitochondrial KATP channel inhibitor 5-HD did not influence GLUT1 translocation during hypoxia. These observations indicate that a decrease of intracellular ATP triggers GLUT1 translocation to the plasma membrane and is mediated by KATP channels, which would contribute to glucose uptake in HUVECs during hypoxia.


Subject(s)
Cell Membrane/metabolism , Deoxyglucose/metabolism , Glucose Transporter Type 1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Adenosine Triphosphate/metabolism , Cell Hypoxia , Cells, Cultured , Glucose Transporter Type 1/genetics , Humans , KATP Channels/genetics , KATP Channels/metabolism , Protein Transport
4.
FASEB J ; 33(6): 7363-7374, 2019 06.
Article in English | MEDLINE | ID: mdl-30865840

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and fatal disease associated with remodeling of the pulmonary artery. We previously reported that the Ca2+-sensing receptor (CaSR) is up-regulated in pulmonary arterial smooth muscle cells (PASMCs) from patients with idiopathic PAH (IPAH) and contributes to enhanced Ca2+ responses and excessive cell proliferation. However, the mechanisms underlying the up-regulation of CaSR have not yet been elucidated. We herein examined involvement of platelet-derived growth factor (PDGF) on CaSR expression, Ca2+ responses, and proliferation in PASMCs. The expression of PDGF receptors was higher in PASMCs from patients with IPAH than in PASMCs from normal subjects. In addition, PDGF-induced activation of PDGF receptors and their downstream molecules [ERK1/2, p38, protein kinase B, and signal transducer and activator of transcription (STAT) 1/3] were sustained longer in PASMCs from patients with IPAH. The PDGF-induced CaSR up-regulation was attenuated by small interfering RNA knockdown of PDGF receptors and STAT1/3, and by the treatment with imatinib. In monocrotaline-induced pulmonary hypertensive rats, the up-regulation of CaSR was reduced by imatinib. The combination of NPS2143 and imatinib additively inhibited the development of pulmonary hypertension. These results suggest that enhanced PDGF signaling is involved in CaSR up-regulation, leading to excessive PASMC proliferation and vascular remodeling in patients with IPAH. The linkage between CaSR and PDGF signals is a novel pathophysiological mechanism contributing to the development of PAH.-Yamamura, A., Nayeem, M. J., Al Mamun, A., Takahashi, R., Hayashi, H., Sato, M. Platelet-derived growth factor up-regulates Ca2+-sensing receptors in idiopathic pulmonary arterial hypertension.


Subject(s)
Gene Expression Regulation/physiology , Hypertension, Pulmonary/physiopathology , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/physiology , Receptors, Calcium-Sensing/biosynthesis , Vascular Remodeling/physiology , Animals , Calcium/physiology , Cell Division/drug effects , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Male , Monocrotaline/toxicity , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Platelet-Derived Growth Factor/pharmacology , Pulmonary Artery/pathology , Rats , Rats, Sprague-Dawley , Receptors, Calcium-Sensing/genetics , Receptors, Platelet-Derived Growth Factor/agonists , Receptors, Platelet-Derived Growth Factor/physiology , Signal Transduction/drug effects , Vascular Remodeling/drug effects
5.
Sci Rep ; 9(1): 1560, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733465

ABSTRACT

Choroidal neovascularization (CNV) is associated with age-related macular degeneration (AMD), a major cause of vision loss among elderly people. Vascular endothelial cell growth factor (VEGF) is essential for the development and progression of AMD, and VEGF signaling molecules are effective targets for the treatment of AMD. We recently reported that activator of G-protein signaling 8 (AGS8), a receptor-independent Gßγ regulator, is involved in VEGF-induced angiogenesis in cultured endothelial cells (EC); however, the role of AGS8 in CNV is not yet understood. This study aimed to explore the role of AGS8 in CNV in cultured cells, explanted choroid tissue, and laser-induced CNV in a mouse AMD model. AGS8 knockdown in cultured choroidal EC inhibited VEGF-induced VEGFR-2 phosphorylation, cell proliferation, and migration. AGS8 knockdown also downregulated cell sprouting from mouse choroidal tissue in ex vivo culture. A mouse model of laser-induced CNV, created to analyze the roles of AGS8 in vivo, demonstrated that AGS8 mRNA was significantly upregulated in choroidal lesions and AGS8 was specifically expressed in the neovasculature. Local AGS8 knockdown in intravitreal tissue significantly inhibited laser-induced AGS8 upregulation and suppressed CNV, suggesting that AGS8 knockdown in the choroid has therapeutic potential for AMD. Together, these results demonstrate that AGS8 plays critical roles in VEGF-induced CNV.


Subject(s)
Choroidal Neovascularization/etiology , Choroidal Neovascularization/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line , Cells, Cultured , Choroidal Neovascularization/pathology , Disease Models, Animal , Disease Susceptibility , Endothelial Cells/metabolism , Gene Expression , Intracellular Signaling Peptides and Proteins/metabolism , Macaca mulatta , Macular Degeneration/etiology , Macular Degeneration/metabolism , Macular Degeneration/pathology , Male , Mice , RNA, Small Interfering/genetics , Vascular Endothelial Growth Factor A/genetics
6.
Exp Cell Res ; 368(1): 13-23, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29649427

ABSTRACT

Vascular endothelial growth factor C (VEGFC) and its cognate receptor VEGFR-3 play a key role in lymphangiogenesis. We previously reported that an ischemia-inducible Gßγ signal regulator, activator of G-protein signaling 8 (AGS8), regulated the subcellular distribution of vascular endothelial growth factor receptor-2 (VEGFR-2) and influenced VEGFA-induced signaling in vascular endothelial cells. Here, we report that AGS8 regulates VEGFR-3, which is another subtype of the VEGF receptor family, and mediates VEGFC signaling in human dermal lymphatic endothelial cells (HDLECs). VEGFC stimulated the proliferation of HDLECs and tube formation by HDLECs, which were inhibited by knocking down AGS8 by small interfering RNA (siRNA). AGS8 siRNA inhibited VEGFC-mediated phosphorylation of VEGFR-3 and its downstream molecules, including ERK1/2 and AKT. Analysis of fluorescence-activated cell sorting and immunofluorescence staining demonstrated that AGS8 knockdown was associated with a reduction of VEGFR-3 at the cell surface. Endocytosis inhibitors did not rescue the decrease of cell-surface VEGFR-3, suggesting that AGS8 regulated the trafficking of VEGFR-3 to the plasma membrane. An immunoprecipitation assay indicated that VEGFR-3 formed a complex including AGS8 and Gßγ in cells. These data suggest the novel regulation of VEGFC-VEGFR-3 by AGS8 in HDLECs and a potential role for AGS8 in lymphangiogenesis.


Subject(s)
Endothelial Cells/metabolism , Neoplasm Proteins/metabolism , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/metabolism , Cell Movement/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Lymphangiogenesis/physiology , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Signal Transduction/physiology
7.
Heliyon ; 3(8): e00369, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28808694

ABSTRACT

AIMS: The present study to address one of the mechanisms in preeclampsia, examined whether levels of oxidative stress, human serum albumin, and endothelial function correlate in pregnant women and whether human serum albumin reduces levels of superoxide produced by NADPH oxidase activation in the human vascular smooth muscle cells. MATERIALS AND METHODS: Pregnant women with (Preeclampsia group, n = 33) and without preeclampsia (Normal group, n = 37) were recruited to determine levels of reactive oxygen species (serum diacron-reactive oxygen metabolite [d-ROM]), and the flow-mediated dilation (FMD). Human coronary arterial smooth muscle cells or omental arteries were subjected to evaluate isometric force recordings, levels of superoxide, western immunoblotting, and immunohistochemistry. The superoxide scavenging assay was also performed in a cell-free system. KEY FINDINGS: Women in the preeclampsia group demonstrated lower FMD and higher serum d-ROM values than those in the normal group. There were the inverse correlations between serum levels of d-ROM and the degree of FMD and between serum levels of albumin and those of d-ROM. D-glucose reduced the levcromakalim-induced dilation of human omental arteries, and it increased levels of superoxide and the recruitment of the NADPH oxidase subunit p47phox in human coronary arterial smooth muscle cells. Human serum albumin (0.05 to 0.5 g/dL) prevented these alterations whereas it exerted no superoxide scavenging effect. SIGNIFICANCE: Serum albumin relates to oxidative stress inversely, but to the endothelial function positively, in pregnant women. Human serum albumin appears to reduce oxidative stress via NADPH oxidase inhibition in the human vascular smooth muscle, indicating that the serum level may be a critical determinant of vascular oxidative stress in some human diseases.

8.
Am J Physiol Cell Physiol ; 311(6): C985-C995, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27834196

ABSTRACT

Hypoxia induces angiogenesis through the VEGF signaling pathway; however, signal propagation of VEGF in hypoxia is not fully understood. In this study, we examined alterations in VEGF signaling during hypoxia conditions and its determinant in endothelial cells. To analyze VEGF signaling during hypoxia, human umbilical vein endothelial cells (HUVECs) were exposed to 3 h of hypoxia (1% O2) followed by 3 h of reoxygenation or 12 h of hypoxia. Hypoxia induced expression of VEGF mRNA, but it was not associated with an increase in tube formation by HUVECs. During 3 h of hypoxia, VEGF-induced phosphorylation of VEGF receptor-2 (VEGFR-2) and downstream molecules were significantly inhibited without a change in VEGFR-2 expression, but it was completely restored after reoxygenation. VEGF-mediated VEGFR-2 phosphorylation is associated with a reduction in cellular ATP in hypoxia conditions (65.93 ± 8.32% of normoxia, means ± SE, P < 0.01). Interestingly, attenuation of VEGFR-2 phosphorylation was restored by addition of ATP to prepared membranes from cells that underwent 3 h of hypoxia. In contrast to 3 h of hypoxia, exposure of cells to 12 h of hypoxia decreased VEGFR-2 expression and VEGF-mediated VEGFR-2 phosphorylation. The magnitude of VEGFR-2 phosphorylation was not fully restored by addition of ATP to prepared membranes from cells exposed to 12 h of hypoxia. These data indicate that ATP is an important determinant of VEGF signaling in hypoxia and suggest that the activation process of VEGFR-2 was modified by sustained hypoxia. These observations contribute to our understanding of signal alterations in VEGF in endothelial cells during hypoxia.


Subject(s)
Adenosine Triphosphate/metabolism , Hypoxia/metabolism , Signal Transduction/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neovascularization, Pathologic , Phosphorylation/physiology , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
PLoS One ; 11(9): e0163151, 2016.
Article in English | MEDLINE | ID: mdl-27632208

ABSTRACT

Sevoflurane exposure impairs the long-term memory in neonates. Whether the exposure to animals in adolescence affects the memory, however, has been unclear. A small hydrolase enzyme of guanosine triphosphate (GTPase) rac1 plays a role in the F-actin dynamics related to the synaptic plasticity, as well as superoxide production via reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activation. The current study was designed to examine whether sevoflurane exposure to mice in early adolescence modifies the long-term learning ability concomitantly with the changes in F-actin constitution as well as superoxide production in the hippocampus according to the levels of rac1 protein expression. Four-week-old mice were subjected to the evaluation of long-term learning ability for three days. On day one, each mouse was allowed to enter a dark chamber for five min to acclimatization. On day two, the procedure was repeated with the addition of an electric shock as soon as a mouse entered the dark chamber. All mice subsequently inhaled 2 L/min air with (Sevoflurane group) and without (Control group) 2.5% sevoflurane for three hours. On day three, each mouse was placed on the platform and retention time, which is the latency to enter the dark chamber, was examined. The brain removed after the behavior test, was used for analyses of immunofluorescence, Western immunoblotting and intracellular levels of superoxide. Sevoflurane exposure significantly prolonged retention time, indicating the enhanced long-term memory. Sevoflurane inhalation augmented F-actin constitution coexisting with the rac1 protein overexpression in the hippocampus whereas it did not alter the levels of superoxide. Sevoflurane exposure to 4-week-old mice accelerates the long-term memory concomitantly with the enhanced F-actin constitution coexisting with the small GTPase rac1 overexpression in the hippocampus. These results suggest that sevoflurane inhalation may amplify long-term memory consolidation via the increased cytoskeleton constitution in the hippocampus of animals in early adolescence.


Subject(s)
Anesthetics, Inhalation/administration & dosage , CA1 Region, Hippocampal/drug effects , GTP Phosphohydrolases/metabolism , Memory, Long-Term/drug effects , Methyl Ethers/administration & dosage , Age Factors , Anesthetics, Inhalation/pharmacology , Animals , Blotting, Western , CA1 Region, Hippocampal/enzymology , Immunohistochemistry , Male , Methyl Ethers/pharmacology , Mice , Mice, Inbred C57BL , Sevoflurane , Superoxides/metabolism
10.
Pflugers Arch ; 468(9): 1555-64, 2016 09.
Article in English | MEDLINE | ID: mdl-27389323

ABSTRACT

Whether high oxygen is harmful to the vascular function is unclear. The present study examined if high oxygen modifies vasodilator effect of cysteine via enhanced oxidative stress and thromboxane production. Rat mesenteric arteries with endothelium at 95 or 50 % oxygen were subjected to isometric force recordings, measurement of thromboxane B2 levels, determination of superoxide and peroxynitrite levels and evaluation of NADPH oxidase subunit protein expression, respectively. L-cysteine (0.01-3 mM) constricted or dilated arteries at 95 and 50 % oxygen, respectively. Thromboxane receptor antagonist SQ-29,548 (1 µM) abolished the constriction at 95 % oxygen. L-cysteine (3 mM) increased levels of thromboxane B2 in arteries upon 95 % oxygen application. L-cysteine relaxed arteries treated with superoxide inhibitor tiron (2 mM) or NADPH oxidase inhibitor gp91ds-tat (1 µM) irrespective of the oxygen concentration while ATP-sensitive K(+) channel inhibitor glibenclamide (1 µM) and cystathionine-γ-lyase (CSE) inhibitor DL-propargylglycine (10 mM) similarly abolished the relaxation. L-cysteine (3 mM) with 95 % oxygen augmented levels of superoxide as well as nitrotyrosine within the artery, concomitantly with enhanced membrane protein expression of NADPH oxidase subunit p47phox. The higher concentration of oxygen attenuates L-cysteine-induced vasodilation via superoxide production mediated by NADPH oxidase along with thromboxane A2 production, resulting in vasoconstriction. The increased levels of superoxide, as well as peroxynitrite, coexist with the impaired vasodilation related to ATP-sensitive K(+) channels and CSE. Higher oxygen with plasma cysteine may cause oxidative stress and vasoconstrictor prostanoid production in blood vessels.


Subject(s)
Cysteine/pharmacology , Mesenteric Arteries/metabolism , Oxidative Stress , Oxygen/pharmacology , Thromboxanes/metabolism , Vasodilation , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Alkynes/pharmacology , Animals , Glyburide/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Glycoproteins/pharmacology , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiology , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Peroxynitrous Acid/metabolism , Rats , Rats, Wistar , Superoxides/metabolism
11.
J Cell Sci ; 129(6): 1210-22, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26826188

ABSTRACT

Activator of G-protein signaling 8 (AGS8, also known as FNDC1) is a receptor-independent accessory protein for the Gßγ subunit, which was isolated from rat heart subjected to repetitive transient ischemia with the substantial development of collaterals. Here, we report the role of AGS8 in vessel formation by endothelial cells. Knockdown of AGS8 by small interfering RNA (siRNA) inhibited vascular endothelial growth factor (VEGF)-induced tube formation, as well as VEGF-stimulated cell growth and migration. VEGF stimulated the phosphorylation of the VEGF receptor-2 (VEGFR-2, also known as KDR), ERK1/2 and p38 MAPK; however, knockdown of AGS8 inhibited these signaling events. Signal alterations by AGS8 siRNA were associated with a decrease of cell surface VEGFR-2 and an increase of VEGFR-2 in the cytosol. Endocytosis blockers did not influence the decrease of VEGFR-2 by AGS8 siRNA, suggesting the involvement of AGS8 in VEGFR-2 trafficking to the plasma membrane. VEGFR-2 formed a complex with AGS8 in cells, and a peptide designed to disrupt AGS8-Gßγ interaction inhibited VEGF-induced tube formation. These data suggest a potential role for AGS8-Gßγ in VEGF signal processing. AGS8 might play a key role in tissue adaptation by regulating angiogenic events.


Subject(s)
Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Rats , Signal Transduction
12.
Proc Natl Acad Sci U S A ; 109(30): 12064-9, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22783020

ABSTRACT

Members of the transforming growth factor-ß superfamily play essential roles in various aspects of embryonic development and physiological organ function. Among them, bone morphogenetic protein (BMP) 9 and BMP10 regulate embryonic vascular development by activating their endothelial receptor ALK1 (activin receptor-like kinase 1, also called Acvrl1). ALK1-mediated intracellular signaling is implicated in the etiologies of human diseases, but their downstream functional proteins are largely unknown. In this study, we identified Tmem100, a gene encoding a previously uncharacterized intracellular transmembrane protein, to be an embryonic endothelium-enriched gene activated by BMP9 and BMP10 through the ALK1 receptor. Tmem100 null mice showed embryonic lethality due to impaired differentiation of arterial endothelium and defects of vascular morphogenesis, which phenocopied most of the vascular abnormalities observed with the Acvrl1/Alk1 deficiency. The activity of Notch- and Akt-mediated signaling, which is essential for vascular development, was down-regulated in Tmem100 null mice. Cre-mediated deletion of Tmem100 in endothelial cells was sufficient to recapitulate the null phenotypes. These data indicated that TMEM100 may play indispensable roles downstream of BMP9/BMP10-ALK1 signaling during endothelial differentiation and vascular morphogenesis.


Subject(s)
Activin Receptors, Type I/metabolism , Arteries/embryology , Cell Differentiation/physiology , Endothelium, Vascular/embryology , Gene Expression Regulation, Developmental/genetics , Membrane Proteins/metabolism , Morphogenesis/physiology , Activin Receptors, Type II , Animals , Arteries/cytology , Blotting, Northern , Blotting, Southern , Blotting, Western , Bone Morphogenetic Proteins/metabolism , Endothelium, Vascular/cytology , Growth Differentiation Factor 2/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Inbred BALB C , Microarray Analysis , Real-Time Polymerase Chain Reaction
13.
Cell Adh Migr ; 3(1): 24-6, 2009.
Article in English | MEDLINE | ID: mdl-19372730

ABSTRACT

Angiogenesis, the growth of new blood vessels from pre-existing vessels, is a process involving endothelial cell proliferation, migration and vascular tube formation. One of the key molecules that regulate this process is the integrin beta3 subunit, a cell adhesion receptor that forms a heterodimer with the integrin alphav subunit and interacts with extracellular matrix components such as fibronectin and vitronectin. Although the integrin beta3 subunit is not normally expressed in quiescent endothelial cells, its expression increases in pathological and physiological angiogenesis, including the vasculature in the ischemic tissues such as tumors. Therefore, the integrin beta3 subunit is known to be a potential target for cancer therapy to block tumor angiogenesis. However, the molecular mechanisms for the transcriptional regulation of this subunit are not fully understood. Recently, we reported that Forkhead transcription factor Foxc2 directly induces expression of the integrin beta3 subunit thorough Forkhead-binding elements in its promoter, thereby regulating integrin beta3-mediated endothelial cell migration and adhesion. Thus, our work now identifies Foxc2 as a novel regulator of angiogenesis. In this commentary, we summarize our new findings and discuss prospects for future research in Foxc2-mediated angiogenesis.


Subject(s)
Forkhead Transcription Factors/metabolism , Integrin beta3/metabolism , Neovascularization, Physiologic , Animals , Cardiovascular System/growth & development , Cardiovascular System/metabolism , Endothelial Cells/metabolism , Humans , Lymphangiogenesis , Models, Biological , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
14.
PLoS One ; 3(6): e2401, 2008 Jun 11.
Article in English | MEDLINE | ID: mdl-18545664

ABSTRACT

BACKGROUND: Recent studies have shown that in the developing embryo, arterial and venous identity is established by genetic mechanisms before circulation begins. Vascular endothelial growth factor (VEGF) signaling and its downstream Notch pathway play critical roles in arterial cell fate determination. We have recently shown that Foxc1 and Foxc2, two closely related Fox transcription factors, are essential for arterial cell specification during development by directly inducing the transcription of Delta-like 4 (Dll4), a ligand for Notch receptors. However, the basic mechanisms whereby the VEGF and Notch signaling pathways control transcriptional regulation of arterial-specific genes have yet to be elucidated. METHODOLOGIES/PRINCIPAL FINDINGS: In the current study, we examined whether and how Foxc transcription factors are involved in VEGF and Notch signaling in induction of Dll4 as well as the Notch target gene Hey2 in endothelial cells. We found that Foxc1 and Foxc2 directly activate the Hey2 promoter via Foxc binding elements. Significantly, Foxc2 physically and functionally interacts with a Notch transcriptional activation complex containing Su(H) and Notch intracellular domain to induce Hey2 promoter activity. Moreover, activation of the Dll4 and Hey2 promoters is induced by VEGF in conjunction with either Foxc1 or Foxc2 more than by either component alone. VEGF-activated PI3K and ERK intracellular pathways modulate the transcriptional activity of Foxc proteins in Dll4 and Hey2 induction. CONCLUSIONS/SIGNIFICANCE: Our new findings demonstrate that Foxc transcriptional factors interact with VEGF and Notch signaling to regulate arterial gene expression in multiple steps of the VEGF-Dll4-Notch-Hey2 signaling pathway.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Endothelium, Vascular/metabolism , Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Receptors, Notch/metabolism , Repressor Proteins/physiology , Signal Transduction , Transcription Factors/physiology , Vascular Endothelial Growth Factor A/metabolism , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , Calcium-Binding Proteins , DNA Primers , Endothelium, Vascular/cytology , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction
15.
J Biol Chem ; 283(35): 23791-800, 2008 Aug 29.
Article in English | MEDLINE | ID: mdl-18579532

ABSTRACT

Forkhead transcription factor Foxc2 is an essential regulator of the cardiovascular system in development and disease. However, the cellular and molecular functions of Foxc2 in vascular endothelial cells are still not fully understood. Here, through gene expression profiling in endothelial cells, we identified molecules associated with cell-extracellular matrix interactions, integrin beta3 (Itgb3), integrin beta5 (Itgb5), and fibronectin, as downstream targets of Foxc2. We found that Itgb3 expression is directly regulated by Foxc2 through multiple Forkhead-binding elements within two high homology regions in the Itgb3 promoter. Because Itgb3 is known to regulate angiogenesis, we further tested whether Foxc2 is directly involved in angiogenesis by regulating Itgb3 expression by in vitro experiments. Overexpression of Foxc2 significantly enhanced endothelial cell migration and adhesion, whereas this effect was strongly inhibited by Itgb3 neutralization antibody. In accordance with these results, pulmonary microvascular endothelial cells isolated from Foxc2 heterozygous mutant mice showed a marked reduction in Itgb3 expression and cell migration. Finally, ex vivo aortic ring assay to test the sprouting and microvessel formation revealed enhanced microvessel outgrowth by Foxc2 overexpression. Conversely, microvessel outgrowth from aortas of Foxc2 heterozygous mutant mice was reduced. Taken together, these results suggest that Foxc2 directly induces Itgb3 expression and regulates angiogenesis by Itgb3-mediated endothelial cell adhesion and migration.


Subject(s)
Endothelial Cells/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/physiology , Integrin beta3/biosynthesis , Neovascularization, Physiologic/physiology , Response Elements/physiology , Animals , Cell Adhesion/physiology , Cell Line , Cell Movement/physiology , Endothelial Cells/cytology , Forkhead Transcription Factors/genetics , Integrin beta Chains/biosynthesis , Integrin beta Chains/genetics , Integrin beta3/genetics , Lung/blood supply , Lung/metabolism , Mice , Mice, Mutant Strains
16.
Biochem Biophys Res Commun ; 367(3): 584-9, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18187037

ABSTRACT

Foxc1 and Foxc2 transcription factors are required for vascular development. However, the molecular mechanisms by which Foxc1 and Foxc2 control angiogenesis, the growth of new blood vessels from pre-existing vessels and capillaries, remain unknown. CXC chemokine ligand 12 (CXCL12) and its receptor, CXCR4, are critical for the process of angiogenesis, including the migration and tube formation of endothelial cells. Here we show that Foxc1 and Foxc2 directly induce CXCR4 expression by activating its promoter in endothelial cells. Furthermore, Foxc1-deficient endothelial cells show a significant reduction in CXCR4 expression as well as CXCL12-stimulated migration. Taken together, these results provide novel evidence that Foxc transcription factors are important regulators of the chemotactic motility of endothelial cells through the induction of CXCR4 expression.


Subject(s)
Cell Movement/drug effects , Chemokine CXCL12/physiology , Endothelial Cells/metabolism , Forkhead Transcription Factors/physiology , Receptors, CXCR4/metabolism , Animals , Cells, Cultured , Chemokine CXCL12/pharmacology , Forkhead Transcription Factors/genetics , Gene Expression Regulation/drug effects , Gene Transfer Techniques , Mice , Mice, Transgenic , Promoter Regions, Genetic/genetics , Receptors, CXCR4/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation/drug effects , Transcriptional Activation/genetics
17.
FEBS Lett ; 536(1-3): 30-4, 2003 Feb 11.
Article in English | MEDLINE | ID: mdl-12586333

ABSTRACT

We previously reported that bone morphogenetic protein (BMP)-2 augments fibroblast growth factor (FGF)-induced neuronal differentiation of PC12 cells by selectively upregulating FGF receptor (FGFR)-1 expression. Here we describe the underlying mechanism. BMP-2 activated Smad proteins in PC12 cells. Overexpression of Smad7 or Smad1, inhibitory and receptor-regulated isoforms, respectively, suppressed or enhanced BMP-2-induced upregulation of FGFR-1 expression. Smad 7 also inhibited the FGF-induced PC12 differentiation. Our findings indicate that activation of a Smad signaling pathway is required for upregulation of FGFR-1 expression by BMP-2 and for the synergistic induction of PC12 differentiation by BMP-2 and FGF.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , DNA-Binding Proteins/physiology , Fibroblast Growth Factors/pharmacology , Neurons/cytology , Trans-Activators/physiology , Transforming Growth Factor beta , Up-Regulation , Animals , Bone Morphogenetic Protein 2 , Cell Differentiation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Drug Synergism , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Rats , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/biosynthesis , Signal Transduction , Smad Proteins , Smad1 Protein , Trans-Activators/antagonists & inhibitors
18.
J Biol Chem ; 278(2): 1303-9, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12417591

ABSTRACT

Mouse embryonic stem-derived cells were recently shown to differentiate into endothelial and smooth muscle cells. In the present study, we investigated whether human umbilical vein endothelium-derived cells retain the potential to differentiate into smooth muscle cells. Examination of biochemical markers, including basic calponin, SM22alpha, prostaglandin E synthase, von Willebrand factor, and PECAM-1, as well as cell contractility, showed that whereas endothelium-derived cells cultured with fibroblast growth factor can be characterized as endothelial cells, when deprived of fibroblast growth factor, a significant fraction differentiates into smooth muscle-like cells. Reapplication of fibroblast growth factor reversed this differentiation. Activin A was up-regulated in fibroblast growth factor-deprived, endothelium-derived cells; moreover, the inhibitory effects of exogenous follistatin and overexpressed Smad7 on smooth muscle-like differentiation confirmed that the differentiation was driven by activin A signaling. These findings indicate that when deprived of fibroblast growth factor, human umbilical vein endothelium-derived cells are capable of differentiating into smooth muscle-like cells through activin A-induced, Smad-dependent signaling, and that maintenance of the endothelial cell phenotype and differentiation into smooth muscle-like cells are reciprocally controlled by fibroblast growth factor-1 and activin A.


Subject(s)
Endothelium, Vascular/cytology , Fibroblast Growth Factor 1/physiology , Muscle, Smooth, Vascular/cytology , Umbilical Veins/cytology , Activins/physiology , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/physiology , Humans , Inhibin-beta Subunits/physiology , Smad7 Protein , Trans-Activators/physiology , Transforming Growth Factor beta/physiology , Transforming Growth Factor beta1 , Vasoconstriction
SELECTION OF CITATIONS
SEARCH DETAIL
...