Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Genes Cells ; 26(10): 782-797, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34333851

ABSTRACT

Bone morphogenetic protein-9 (BMP9), a member of the transforming growth factor ß (TGFß) superfamily, plays important roles in the development and maintenance of various cell lineages via complexes of type I and type II TGFß receptors. Endoglin is a coreceptor for several TGFß family members, including BMP9, which is highly expressed in a particular stage of differentiation in erythroid cells as well as in endothelial cells. Although the importance of the interaction between BMP9 and endoglin for endothelial development has been reported, the contribution of BMP9 to endoglin-expressing erythroid cells remains to be clarified. To address this point, we prepared an anti-BMP9 antibody that blocks the BMP9-endoglin interaction. Of note, challenge with the antibody promotes erythropoiesis in wild-type mice but not in a mouse model of renal anemia in which erythropoietin (EPO) production in the kidneys is genetically ablated. While endoglin-positive erythroid progenitors are mainly maintained as progenitors when bone marrow-derived lineage-negative and cKit-positive cells are cultured in the presence of EPO and stem cell factor, the erythroid-biased accumulation of progenitors is impeded by the presence of BMP9. Our findings uncover an unrecognized role for BMP9 in attenuating erythroid differentiation via its interaction with endoglin on erythroid progenitors.


Subject(s)
Erythropoiesis , Growth Differentiation Factor 2 , Animals , Endoglin/genetics , Endothelial Cells , Erythroid Precursor Cells , Growth Differentiation Factor 2/genetics , Mice , Signal Transduction
2.
Int J Cancer ; 134(8): 1785-95, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24155219

ABSTRACT

Cancer-associated fibroblasts (CAFs) have recently been implicated in tumor growth and metastasis in gastric cancer. Cancer stem cells (CSCs) have been proposed to have an important role in cancer progression. The aim of this study was to clarify the effect of CAFs on CSCs characteristics in gastric carcinoma. Scirrhous gastric cancer cell lines, OCUM-12 and OCUM-2MD3, and non-scirrhous gastric cancer cell lines, MKN-45 and MKN-74, were used. OCUM-12/side population (SP) cells and OCUM-2MD3/SP cells were sorted by flow cytometry as CSC-rich cells from the parent cells. CaF-37 was established from the tumoral gastric specimens as CAFs. Flow cytometric analysis of SP fraction, spheroid colony assay, and RT-PCR analysis of CSC markers were performed to identify CSCs properties. Effect of CAFs on the tumorigenicity by OCUM-12/SP cells was examined using nude mice. CAF CM significantly increased the percentages of the SP fraction of OCUM-12/SP and OCUM-2MD3/SP cells, but not that of MKN-45/SP and MKN-74/SP cells. Taken together, CM from CaF-37 significantly increased the number of spheroid colonies and the expression level of CSC markers of OCUM-12/SP and OCUM-2MD3/SP cells. These stimulating-activities by CM were significantly decreased by TGFß inhibitors, but not FGFR and cMet inhibitor. Tumorigenicity by subcutaneous coinoculation of OCUM-12/SP cells with CAFs was significantly high in comparison with that by OCUM-12/SP cells alone. Phospho-Smad2 expression level was significantly increased by co-inoculation with CAFs. These findings suggested that CAFs might regulate the stemness of CSCs in scirrhous gastric cancer by TGFß signaling.


Subject(s)
Adenocarcinoma, Scirrhous/metabolism , Fibroblasts/metabolism , Neoplastic Stem Cells/metabolism , Stomach Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment , Actins/biosynthesis , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/biosynthesis , Smad2 Protein/metabolism , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
3.
PLoS One ; 8(5): e62310, 2013.
Article in English | MEDLINE | ID: mdl-23690936

ABSTRACT

Epithelial mesenchymal transition (EMT) is considered to be correlated with malignancy of cancer cells and responsible for cancer invasion and metastasis. We previously reported that distant metastasis was associated with hypoxia in gastric cancer. We therefore investigated the effect of hypoxic condition on EMT of gastric cancer cells. Gastric cancer cells were cultured in normoxia (21% O2) or hypoxia (1% O2) for 24 h. EMT was evaluated as the percentage of spindle-shaped cells in total cells. Effect of transforming growth factor ß1 (TGFß1) or tyrosine kinase inhibitors on the EMT was evaluated. The expression level of TGFß1 and TGFßR was evaluated by real time RT-PCR. The TGFß1 production from cancer cells was measured by ELISA. Hypoxia stimulated EMT of OCUM-2MD3 and OCUM-12 cells, but not that of OCUM-2M cells. The expression level of TGFß1 mRNA under hypoxia was significantly higher than that under normoxia in all of three cell lines. The expression level of TGFßR mRNA was significantly increased by hypoxia in OCUM-2MD3 cells, but not in OCUM-2M cells. TGFßR inhibitor, SB431542 or Ki26894, significantly suppressed EMT of OCUM-2MD3 and OCUM-12. TGFß1 production from OCUM-2MD3 and OCUM-12 cells was significantly increased under hypoxia in comparison with that under normoxia. These findings might suggest that hypoxia stimulates the EMT of gastric cancer cells via autocrine TGFß/TGFßR signaling.


Subject(s)
Autocrine Communication/physiology , Cell Hypoxia/physiology , Epithelial-Mesenchymal Transition/physiology , Receptors, Transforming Growth Factor beta/metabolism , Stomach Neoplasms/physiopathology , Transforming Growth Factor beta1/metabolism , Analysis of Variance , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
4.
Lab Invest ; 92(8): 1100-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22584670

ABSTRACT

Skeletal muscle expressing Pro104Leu mutant caveolin 3 (CAV3(P104L)) in mouse becomes atrophied and serves as a model of autosomal dominant limb-girdle muscular dystrophy 1C. We previously found that caveolin 3-deficient muscles showed activated intramuscular transforming growth factor beta (TGF-ß) signals. However, the cellular mechanism by which loss of caveolin 3 leads to muscle atrophy is unknown. Recently, several small-molecule inhibitors of TGF-ß type I receptor (TßRI) kinase have been developed as molecular-targeting drugs for cancer therapy by suppressing intracellular TGF-ß1, -ß2, and -ß3 signaling. Here, we show that a TßRI kinase inhibitor, Ki26894, restores impaired myoblast differentiation in vitro caused by activin, myostatin, and TGF-ß1, as well as CAV3(P104L). Oral administration of Ki26894 increased muscle mass and strength in vivo in wild-type mice, and improved muscle atrophy and weakness in the CAV3(P104L) mice. The inhibitor restored the number of satellite cells, the resident stem cells of adult skeletal muscle, with suppression of the increased phosphorylation of Smad2, an effector, and the upregulation of p21 (also known as Cdkn1a), a target gene of the TGF-ß family members in muscle. These data indicate that both TGF-ß-dependent reduction in satellite cells and impairment of myoblast differentiation contribute to the cellular mechanism underlying caveolin 3-deficient muscle atrophy. TßRI kinase inhibitors could antagonize the activation of intramuscular anti-myogenic TGF-ß signals, thereby providing a novel therapeutic rationale for the alternative use of this type of anticancer drug in reversing muscle atrophy in various clinical settings.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Caveolin 3/deficiency , Muscular Dystrophies, Limb-Girdle/drug therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Activin Receptors, Type I/pharmacology , Activins/metabolism , Activins/pharmacology , Animals , Caveolin 3/genetics , Caveolin 3/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Disease Models, Animal , Female , Gene Expression Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Transgenic , Muscle Development/drug effects , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Myoblasts/pathology , Myostatin/metabolism , Myostatin/pharmacology , Receptor, Transforming Growth Factor-beta Type I , Satellite Cells, Skeletal Muscle/drug effects , Signal Transduction , Transforming Growth Factor beta1/genetics
5.
J Clin Biochem Nutr ; 48(2): 134-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21373266

ABSTRACT

Lipopolysaccharide-stimulated leukocytes secrete proinflammatory cytokines including tumor necrosis factor-α and interleukin-12. Over-activation of host defense systems may result in severe tissue damage and requires regulation. Granulocyte colony-stimulating factor and interleukin-10 are candidate cytokines for inducing tolerance to lipopolysaccharide re-stimulation. We compared cytokines secreted by lipopolysaccharide-stimulated blood cells from patients who had survived gram negative bacterial pneumonia (Pseudomonas aeruginosa, Escherichia coli or Proteus mirabilis, n = 26) and age-matched healthy volunteers (n = 18). Interleukin-12p70 and tumor necrosis factor-α expression was significantly lower in patients (p = 0.0039 and p<0.001) compared to healthy controls, while granulocyte colony-stimulating factor production was markedly higher in patients (p<0.001). Levels of interleukin-10 were comparable. Granulocyte colony-stimulating factor expression was inversely correlated with interleukin-12p70 (R = -0.71, p<0.001) and tumor necrosis factor-α (R = -0.64, p<0.001) expression; interleukin-10 showed no significant correlation. In unstimulated leukocytes from patients, cAMP levels were significantly raised (p = 0.020) and were correlated inversely with interleukin-12p70 levels (R = -0.81, p<0.001) and directly with granulocyte colony-stimulating factor (R = 0.72, p = 0.0020), matrix metalloproteinase-9 (R = 0.67, p = 0.0067) and interleukin-10 (R = 0.54, p = 0.039) levels. Our results demonstrate that granulocyte colony-stimulating factor production by lipopolysaccharide-stimulated leukocytes is a useful indicator of tolerance induction in surviving pneumonia patients and that measuring cAMP in freshly isolated leukocytes may also be clinically significant.

6.
Cancer Sci ; 101(8): 1846-52, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20518790

ABSTRACT

Transforming growth factor-beta (TGF-beta) signals are closely associated with the distant metastases of gastric cancer. The aim of this study was to clarify the effect of a TGF-beta receptor I (TbetaR-I) phosphorylation inhibitor, Ki26894, in combination with anticancer drugs, on the lymph node (LN) metastasis of scirrhous gastric cancer. A novel TbetaR-I kinase inhibitor, Ki26894, inhibits the phosphorylation of Smad2 at the ATP binding site of TbetaR-I. S1 is a 5-fluorouracil analog. The human scirrhous gastric cancer cell line OCUM-2MLN and the human gastric fibroblasts NF-33 were used. OCUM-2MLM cells in the upper well and NF-33 cells in the lower well were co-incubated with or without Ki26894. The proliferation of OCUM-2MLN cells was significantly stimulated by co-culture with NF-33 cells. Ki26894 significantly suppressed the growth interactions between OCUM-2MLN cells and NF-33 cells. Gastric cancer models established by orthotopic inoculation of OCUM-2MLN cells showed diffusely infiltrating gastric adenocarcinoma accompanied by LN metastases. We divided these mice into four groups, (control vehicle, Ki26894, S1, Ki26894 plus S1), and examined the effect of Ki26894 and/or S1 on phosphorylation of Smad2, tumor size, LN metastases, and lymphatic involvements. Ki26894 inhibited the Smad2 phosphorylation of cancer cells and decreased the extent of lymphatic involvement, compared with the control or S1 only group. The Ki26894 plus S1 administration group significantly suppressed tumor growth and decreased LN metastasis more effectively than either alone. These findings suggested that the TbetaR-I kinase inhibitor with S1 is useful for the treatment of scirrhous gastric carcinoma with LN metastasis. (Cancer Sci 2010).


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Oxonic Acid/administration & dosage , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Stomach Neoplasms/drug therapy , Tegafur/administration & dosage , Activin Receptors, Type I/administration & dosage , Animals , Drug Combinations , Humans , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Phosphorylation , Receptor, Transforming Growth Factor-beta Type I , Smad2 Protein/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta1/pharmacology
8.
Mol Ecol Resour ; 9(3): 819-21, 2009 May.
Article in English | MEDLINE | ID: mdl-21564755

ABSTRACT

We developed 12 polymorphic microsatellite loci for the Asian honeybee, Apis cerana using the magnetic particle method. Eight of these 12 were highly polymorphic, having four to seven alleles with an expected heterozygosity of 0.38 to 0.78. The primers also produce polymorphic products in related honeybee species such as Apis nigrocincta. These loci can be used to study parameters associated with genetic structure, such as paternity frequency and worker reproduction.

9.
J Med Chem ; 51(11): 3326-9, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18479116

ABSTRACT

We synthesized and evaluated various [2-(4-quinolyloxy)phenyl]methanone derivatives. These compounds had novel chemical structures that were distinct from those of previously reported inhibitors. Biological data suggested that these compounds inhibited transforming growth factor-beta signaling by interacting with the ATP-binding pocket of the transforming growth factor-beta type I receptor kinase domain. Here, we report on the synthesis and structure-activity relationships of the compounds in this series.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Quinolines/chemical synthesis , Transforming Growth Factor beta/metabolism , Activin Receptors, Type I/metabolism , Cell Line, Tumor , Genes, Reporter , Humans , Luciferases/biosynthesis , Luciferases/genetics , Phosphorylation , Quinolines/chemistry , Quinolines/pharmacology , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Structure-Activity Relationship , Transforming Growth Factor beta/pharmacology
10.
Cancer Sci ; 98(1): 127-33, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17129361

ABSTRACT

Transforming growth factor (TGF)-beta signaling has been shown to promote tumor growth and metastasis in advanced cancer. Use of inhibitors of TGF-beta signaling may thus be a novel strategy for treatment of patients with such cancers. In this study, we investigated the effects of a novel TGF-beta type I receptor (TbetaR-I) kinase inhibitor, Ki26894, on bone metastasis of a highly bone-metastatic variant of human breast cancer MDA-MB-231 cells, termed MDA-MB-231-5a-D (MDA-231-D). Ki26894 blocked TGF-beta signaling in MDA-231-D cells, as detected by suppression of phosphorylation of Smad2 and inhibition of TGF-beta-responsive reporter activity. Moreover, Ki26894 decreased the motility and the invasion of MDA-231-D cells induced by TGF-beta in vitro. Ki26894 also suppressed transcription of plasminogen activator inhibitor-1 (PAI-1), parathyroid hormone-related protein (PTHrP), and interleukin-11 (IL-11) mRNA of MDA-231-D cells, which were stimulated by TGF-beta. X-ray radiography revealed that systemic Ki26894 treatment initiated 1 day before the inoculation of MDA-231-D cells into the left ventricle of BALB/cnu/nu female mice resulted in decreased bone metastasis of breast cancer cells. Moreover, Ki26894 prolonged the survival of mice inoculated with MDA-231-D cells compared to vehicle-treated mice. These findings suggest that TbetaR-I kinase inhibitors such as Ki26894 may be useful for blocking the progression of advanced cancers.


Subject(s)
Activin Receptors, Type I/pharmacokinetics , Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/pathology , Neoplasm Metastasis/prevention & control , Protein Kinase Inhibitors/pharmacology , Activin Receptors, Type I/drug effects , Animals , Bone Neoplasms/secondary , Female , Humans , Immunoblotting , In Vitro Techniques , Mice , Neoplasm Invasiveness/prevention & control , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/drug effects , Reverse Transcriptase Polymerase Chain Reaction
11.
Am J Physiol Regul Integr Comp Physiol ; 290(2): R322-30, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16166203

ABSTRACT

We tested the hypothesis that renal sympathetic nerve activity (RSNA) to the ischemic and nonischemic regions responded differently during partial ischemia of the kidney in pentobarbital-anesthetized cats. The renal artery divides into two branches at the front of the renal hilus: one branch perfuses predominantly the dorsal half of the kidney, and the other perfuses its ventral half. We identified the innervated area of a renal nerve bundle by supramaximal electrical stimulation and subsequently determined the changes in RSNA in response to occlusion of either renal arterial branch for 3 min. RSNA to the nonischemic region of the kidney gradually decreased by 23 +/- 4% during partial renal ischemia, whereas RSNA to the ischemic region of the same kidney showed no significant change. Crushing either all renal nerve bundles or only the renal nerve bundles terminated to the ischemic region abolished the decrease in RSNA to the nonischemic region. Furthermore, intra-arterial administration of a prostaglandin synthesis inhibitor (meclofenamate, 4 mg/kg) abolished the decrease in RSNA to the nonischemic region of the kidney. Following spinal transection at the level of T7, the inhibitory response in RSNA to the nonischemic region disappeared, whereas the RSNA to the ischemic region was markedly augmented by 47 +/- 17%. Thus it is likely that renal chemoreceptors activated during renal partial ischemia elicit heterogeneous control of renal sympathetic outflows to the ischemic and nonischemic regions of the same kidney, which may be determined by a net output between the supraspinal inhibitory and spinal excitatory reflexes.


Subject(s)
Adrenergic Fibers/physiology , Ischemia/physiopathology , Kidney/blood supply , Kidney/innervation , Animals , Blood Pressure , Cats , Heart Rate , Kidney/drug effects , Meclofenamic Acid/pharmacology , Prostaglandin Antagonists/pharmacology
12.
Exp Neurol ; 199(2): 274-80, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16337192

ABSTRACT

This study was performed in order to evaluate the angiogenic effect of implantation of either peripheral blood mononuclear cells (PBMNCs) or bone marrow mononuclear cells (BMMNCs) on diabetic peripheral neuropathy. Streptozotocin (50 mg/kg) was injected intravenously into 6-week-old male Lewis rats. Four weeks after the induction of diabetes, 6 x 10(7) of PBMNCs or 1 x 10(8) of BMMNCs were implanted into the left hindlimb muscle. Motor nerve conduction velocity (MNCV) was monitored before and after implantation. At the end of the experiment, bilateral nerve blood flow (NBF) was measured by laser Doppler and the number of vessels in the sciatic nerves quantified by Factor VIII staining of the sections. Diabetes resulted in an approximately 20% reduction (P < 0.01) in sciatic MNCV. Four weeks after implantation, MNCV was improved by 54% with PBMNCs and by 67% with BMMNCs (both P < 0.01). Moreover, the effects of implantation were almost abolished by administration of VEGF-neutralizing antibody. Sciatic NBF was reduced by approximately 50% by diabetes (P < 0.05). This reduction in perfusion was improved by 74% by implantation of PBMNCs and by 62% by implantation of BMMNCs (P < 0.05 and P < 0.01, respectively). These effects were observed only in the implanted limb. Immunohistochemical staining of sciatic nerve sections for Factor VIII showed no significant increase in the number of vessels in the sciatic nerve following implantation of either PBMNCs or BMMNCs. These data suggest that implantation of hematopoietic mononuclear cell fractions is associated with an improvement in MNCV as a result of arteriogenic effects in the sciatic nerve, and that VEGF may contribute to this effect. This improvement occurred in the absence of angiogenesis. Implantation of these cell fractions may therefore be a potential new therapeutic method for treating diabetic peripheral neuropathy.


Subject(s)
Diabetes Mellitus, Experimental/surgery , Diabetic Neuropathies/surgery , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Analysis of Variance , Animals , Antibodies/administration & dosage , Antigens, CD34/metabolism , Diabetes Mellitus, Experimental/complications , Diabetic Neuropathies/etiology , Diabetic Neuropathies/physiopathology , Disease Models, Animal , Immunohistochemistry/methods , Laser-Doppler Flowmetry/methods , Male , Neural Conduction/drug effects , Neural Conduction/physiology , Nitric Oxide Synthase Type III/metabolism , Rats , Rats, Inbred Lew , Regional Blood Flow/physiology , Time Factors , Vascular Endothelial Growth Factor A/administration & dosage , Vascular Endothelial Growth Factor A/immunology
13.
Cancer Sci ; 96(5): 274-82, 2005 May.
Article in English | MEDLINE | ID: mdl-15904468

ABSTRACT

The Notch proteins constitute a family of transmembrane receptors that play a pivotal role in cellular differentiation, proliferation and apoptosis. Although it has been recognized that excess Notch signaling is potentially tumorigenic, little is known about precise mechanisms through which dysregulated Notch signaling induces neoplastic transformation. Here we demonstrate that Notch signaling has a transcriptional cross-talk with transforming growth factor-beta (TGF-beta) signaling, which is well characterized by its antiproliferative effects. TGF-beta-mediated transcriptional responses are suppressed by constitutively active Notch1, and this inhibitory effect is canceled by introduction of transcriptional coactivator p300. We further show that this blockade of TGF-beta signaling is executed by the sequestration of p300 from Smad3. Moreover, in a human cervical carcinoma cell line, CaSki, in which Notch1 is spontaneously activated, suppression of Notch1 expression with small interfering RNA significantly restores the responsiveness to TGF-beta. Taken together, we propose that Notch oncoproteins promote cell growth and cancer development partly by suppressing the growth inhibitory effects of TGF-beta through sequestrating p300 from Smad3.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Receptors, Cell Surface/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Chlorocebus aethiops , DNA-Binding Proteins/genetics , E1A-Associated p300 Protein , Humans , Mice , Mutation/genetics , Nuclear Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Notch1 , Receptors, Cell Surface/genetics , Signal Transduction/drug effects , Smad Proteins , Trans-Activators/genetics , Transcription Factors/genetics , Transcription, Genetic/drug effects
14.
Oncogene ; 23(5): 1043-51, 2004 Feb 05.
Article in English | MEDLINE | ID: mdl-14762439

ABSTRACT

The transforming growth factor-beta (TGF-beta)-Smad signaling pathway inhibits the growth of human epithelial cells and plays a role in tumor suppression. The Smad4 gene is mutated or deleted in 50% of pancreatic cancers. In this study, the Smad4-null pancreatic cancer cell line BxPC-3 was transfected with either the Smad4 expression vector or the empty vector and incubated in the presence or absence of TGF-beta. The cells were analysed using a cDNA microarray, which included 2280 named genes to screen for target genes regulated by TGF-beta in either a Smad4-dependent or -independent manner. The microarray and subsequent quantitative RT-PCR analysis demonstrated that the Smad4-independent and -dependent signaling pathways driven by TGF-beta upregulated only one of the 2280 genes, respectively, suggesting that Smad4-independent signaling downstream of TGF-beta might be as widespread as Smad4-dependent signaling. In this study, we demonstrated that the cyclin-dependent kinase inhibitor p21/WAF1, which has been considered the major effector of the Smad-dependent growth inhibitory signal of TGF-beta, is upregulated in a Smad4-independent manner. The upregulation occurs through Smad2/3-dependent transcriptional activation of the p21/WAF1 promoter region. These results suggest a novel mechanism of gene regulation, that is, a novel signal mediator other than Smad4.


Subject(s)
Cyclins/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology , Active Transport, Cell Nucleus/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/drug effects , Cyclins/genetics , DNA, Complementary/analysis , DNA-Binding Proteins/metabolism , Endoribonucleases , Enzyme Induction , Gene Deletion , Gene Expression Regulation/drug effects , Genes, Tumor Suppressor , Humans , Keratinocytes/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , RNA, Messenger/metabolism , Signal Transduction , Smad2 Protein , Smad3 Protein , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcriptional Activation
15.
Proc Natl Acad Sci U S A ; 100(26): 15577-82, 2003 Dec 23.
Article in English | MEDLINE | ID: mdl-14671321

ABSTRACT

In the transforming growth factor beta (TGF-beta) superfamily, activin A, TGF-beta1, and bone morphogenic protein 4 (BMP-4) have various effects on hematopoiesis, including early mesodermo-hematogenesis. After these cytokines bind to their respective receptor, a regulatory Smad is phosphorylated and becomes associated with Smad4, the common Smad, and the resulting complex translocates to the nucleus to regulate transcription. DLX1 is the product of a member of the distal-less homeobox gene family, which is known to have important roles in embryogenesis, particularly in craniofacial development, and in GABAergic neurogenesis. DLX1 has been reported to be temporally and spatially coexpressed with BMP-4 during embryogenesis in selected contexts. We report here that, in addition to the previously reported regions/cells, DLX1 is expressed in hematopoietic cells in a lineage-dependent manner and that DLX1 interacts with Smad4 through its homeodomain. We show that it blocks multiple signals from TGF-beta superfamily cytokines such as activin A, TGF-beta1, and BMP-4, including differentiation of a hematopoietic cell line by activin A. Taken together, these data suggest that DLX1 may function as a regulator of multiple signals from TGF-beta superfamily members in broad biological contexts during blood production.


Subject(s)
Activins/metabolism , DNA-Binding Proteins/metabolism , Homeodomain Proteins/metabolism , Inhibin-beta Subunits/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , DNA, Complementary , DNA-Binding Proteins/genetics , Gene Library , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/genetics , Humans , Leukemia, Erythroblastic, Acute , Molecular Sequence Data , Smad4 Protein , Trans-Activators/genetics , Transcription Factors , Tumor Cells, Cultured
16.
J Cell Biol ; 163(6): 1303-11, 2003 Dec 22.
Article in English | MEDLINE | ID: mdl-14676305

ABSTRACT

Recent findings have shown that embryonic vascular progenitor cells are capable of differentiating into mural and endothelial cells. However, the molecular mechanisms that regulate their differentiation, proliferation, and endothelial sheet formation remain to be elucidated. Here, we show that members of the transforming growth factor (TGF)-beta superfamily play important roles during differentiation of vascular progenitor cells derived from mouse embryonic stem cells (ESCs) and from 8.5-days postcoitum embryos. TGF-beta and activin inhibited proliferation and sheet formation of endothelial cells. Interestingly, SB-431542, a synthetic molecule that inhibits the kinases of receptors for TGF-beta and activin, facilitated proliferation and sheet formation of ESC-derived endothelial cells. Moreover, SB-431542 up-regulated the expression of claudin-5, an endothelial specific component of tight junctions. These results suggest that endogenous TGF-beta/activin signals play important roles in regulating vascular growth and permeability.


Subject(s)
Benzamides/pharmacology , Blood Vessels/embryology , Dioxoles/pharmacology , Endothelium, Vascular/metabolism , Neovascularization, Physiologic/physiology , Pluripotent Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Activins/metabolism , Activins/pharmacology , Animals , Blood Vessels/cytology , Blood Vessels/metabolism , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Claudin-5 , Endothelium, Vascular/cytology , Fetus , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Mice , Neovascularization, Physiologic/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tight Junctions/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
17.
Cancer Res ; 63(22): 7791-8, 2003 Nov 15.
Article in English | MEDLINE | ID: mdl-14633705

ABSTRACT

Transforming growth factor-beta (TGF-beta) has growth-stimulating effects on mesenchymal cells and several tumor cell lines. The signaling pathway for this effect is, however, not well understood. We examined how TGF-beta stimulates proliferation of MG63 human osteosarcoma cells. Two distinct type I receptors for TGF-beta, ALK-1 and ALK-5, were expressed and functional in MG63 cells. Of these two receptors, ALK-5 appears to be responsible for the growth stimulation because expression of constitutively active ALK-5, but not ALK-1, stimulated proliferation of MG63 cells. SB-431542 (0.3 microM), a novel inhibitor of ALK4/5/7 kinase, suppressed TGF-beta-induced growth stimulation. DNA microarray analysis as well as quantitative real-time PCR analysis of RNAs from TGF-beta-treated cells demonstrated that several growth factors, including platelet-derived growth factor AA, were induced in response to TGF-beta in MG63 cells. Gleevec (1 microM) as well as AG1296 (5 microM) inhibited TGF-beta-induced growth stimulation of MG63 cells, suggesting that platelet-derived growth factor AA was mainly responsible for the growth-stimulatory effect of TGF-beta. We also examined the mechanisms of perturbation of growth-suppressing signaling in MG63 cells. We found that expression of c-Myc, which is down-regulated by TGF-beta in many other cells, was up-regulated in MG63 cells, suggesting that up-regulation of c-Myc expression may be the mechanism canceling growth-suppressing signaling of TGF-beta in MG63 cells.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Bone Neoplasms/pathology , Dioxoles/pharmacology , Osteosarcoma/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Activin Receptors, Type I/biosynthesis , Activin Receptors, Type I/physiology , Activin Receptors, Type II , Animals , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/biosynthesis , Cyclins/genetics , Drug Interactions , Gene Expression Regulation, Neoplastic/drug effects , Humans , Imatinib Mesylate , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/biosynthesis , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology , Up-Regulation/drug effects
18.
Jpn J Physiol ; 53(2): 97-103, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12877766

ABSTRACT

The aim of this study was to examine whether or not stimulation of the hypothalamic defense area is capable of inducing sympathetic vasodilatation of the forelimb vascular bed in anesthetized cats. When the hypothalamic defense area was electrically stimulated, brachial blood flow velocity (brachial BFV) and vascular conductance were increased as well as femoral BFV and vascular conductance. Brachial BFV and vascular conductance increased by 110-139% during hypothalamic stimulation. These increases were blunted to approximately one-fifth of the control responses following i.v. injection of a synthesis inhibitor of nitric oxide, N(omega)-nitro-L-arginine methyl ester (L-NAME). The attenuating effect of L-NAME on forelimb vasodilatation evoked by hypothalamic stimulation was greater than that on hindlimb vasodilatation. The combined administration of L-NAME and atropine sulfate eliminated nearly all of the increases in brachial BFV and vascular conductance during hypothalamic stimulation. From the present results, we conclude that stimulation of the hypothalamic defense area is able to induce neurogenic vasodilatation of the cat forelimb vascular bed, which is greatly mediated with a nitric oxide mechanism. The contribution of nitric oxide to neurogenic vasodilatation seems to be greater in the forelimbs than hindlimbs.


Subject(s)
Forelimb/physiology , Hypothalamus/physiology , Nitric Oxide/metabolism , Vasodilation/physiology , Anesthesia , Animals , Aorta/physiology , Atropine/pharmacology , Blood Flow Velocity/drug effects , Blood Pressure Determination , Brachial Artery/physiology , Cardiac Output/physiology , Carotid Arteries/physiology , Cats , Data Interpretation, Statistical , Electric Stimulation , Enzyme Inhibitors/pharmacology , Femoral Artery/physiology , Heart Rate/physiology , Hypothalamus/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Parasympatholytics/pharmacology , Respiration, Artificial , Sympathetic Nervous System/physiology , Tracheotomy , Vasodilation/drug effects
19.
Immunity ; 18(5): 675-85, 2003 May.
Article in English | MEDLINE | ID: mdl-12753744

ABSTRACT

The Notch genes play a key role in cellular differentiation. The significance of Notch1 during thymocyte development is well characterized, but the function of Notch2 is poorly understood. Here we demonstrate that Notch2 but no other Notch family member is preferentially expressed in mature B cells and that conditionally targeted deletion of Notch2 results in the defect of marginal zone B (MZB) cells and their presumed precursors, CD1d(hi) fraction of type 2 transitional B cells. Among Notch target genes, the expression level of Deltex1 is prominent in MZB cells and strictly dependent on that of Notch2, suggesting that Deltex1 may play a role in MZB cell differentiation.


Subject(s)
B-Lymphocytes/metabolism , Carrier Proteins , Cell Differentiation/genetics , Cell Lineage/genetics , Receptors, Cell Surface/genetics , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Heterozygote , Mice , Mice, Knockout , Proteins/metabolism , Receptor, Notch2 , Receptors, Cell Surface/biosynthesis
20.
Immunity ; 18(5): 699-711, 2003 May.
Article in English | MEDLINE | ID: mdl-12753746

ABSTRACT

Hematopoietic stem cells (HSCs) are thought to arise in the aorta-gonad-mesonephros (AGM) region of embryo proper, although HSC activity can be detected in yolk sac (YS) and paraaortic splanchnopleura (P-Sp) when transplanted in newborn mice. We examined the role of Notch signaling in embryonic hematopoiesis. The activity of colony-forming cells in the YS from Notch1(-/-) embryos was comparable to that of wild-type embryos. However, in vitro and in vivo definitive hematopoietic activities from YS and P-Sp were severely impaired in Notch1(-/-) embryos. The population representing hemogenic endothelial cells, however, did not decrease. In contrast, Notch2(-/-) embryos showed no hematopoietic deficiency. These data indicate that Notch1, but not Notch2, is essential for generating hematopoietic stem cells from endothelial cells.


Subject(s)
Cell Differentiation/physiology , Hematopoietic Stem Cells/metabolism , Membrane Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors , Animals , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/cytology , Mice , Receptor, Notch1 , Receptor, Notch2 , Yolk Sac/cytology , Yolk Sac/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL