Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
Add more filters










Publication year range
1.
Oncogenesis ; 5(9): e259, 2016 Sep 12.
Article in English | MEDLINE | ID: mdl-27617643

ABSTRACT

Onset of the cancer mesenchymal program is closely associated with cancer malignancy and drug resistance. Among the different epithelial-mesenchymal transition (EMT)-associated transcriptional factors, ZEB1 has a key role in inducing the mesenchymal phenotypes and stem cell-like properties of different breast cancer cells. ARF6 and its effector AMAP1 are frequently overexpressed in breast cancer cells, and promote invasion, metastasis and drug resistance. EPB41L5 is induced during EMT, and mediates the disruption of E-cadherin-based cell-cell adhesion and the promotion of focal adhesion dynamics. Here we show that EPB41L5 is an integral component of the ARF6-based pathway, which is induced by ZEB1. We found that EPB41L5 is expressed at high levels in malignant breast cancer cells and binds to AMAP1. ZEB1 induced EPB41L5 both in cancer cells and normal cells. This relationship was recaptured with The Cancer Genome Atlas RNASeq data set, and correlated with the poor outcome of the patients. In contrast, diversified events, such as tumor growth factor ß1 stimulation, expression of SNAI1 and TP53 mutation, can each cause the induction of ZEB1 and EPB41L5, depending on the cellular context. Our results demonstrated that the ZEB1-EPB41L5 axis is at the core of the cancer mesenchymal program that drives ARF6-based invasion, metastasis and drug resistance of significant populations of primary breast cancers, and is tightly correlated with the poor outcomes of patients.

2.
Neuroscience ; 151(4): 1225-35, 2008 Feb 19.
Article in English | MEDLINE | ID: mdl-18201830

ABSTRACT

Calcium entry into the postsynaptic neuron through N-methyl-D-aspartate-type glutamate receptors (NMDARs) triggers the induction of long-term potentiation (LTP), which is considered to contribute to synaptic plasticity and plays a critical role in behavioral learning. We report here that activin, a member of the transforming growth factor-beta (TGF-beta) superfamily, promotes phosphorylation of NMDARs and increases the Ca2+ influx through these receptors in primary cultured rat hippocampal neurons. This signal transduction occurs in a functional complex of activin receptors, NMDARs, and Src family tyrosine kinases, including Fyn, formed on a multimer of postsynaptic scaffolding postsynaptic density protein 95/Dlg/ZO-1 (PDZ), activin receptor interacting protein 1 (ARIP1). Activin-induced NMDAR activation persists for more than 24 h, which is complimentary to the activation time of NMDARs by brain-derived neurotrophic factor (BDNF). Our results suggest that activin is a unique and powerful potentiator for NMDAR-dependent signaling, which could be involved in the regulatory mechanisms of synaptic plasticity.


Subject(s)
Activins/pharmacology , Neurons/drug effects , Proteins/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Adaptor Proteins, Signal Transducing , Animals , Benzamides/pharmacology , Calcium/metabolism , Cells, Cultured , Dioxoles/pharmacology , Dizocilpine Maleate/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/pharmacology , Guanylate Kinases , Hippocampus/cytology , Immunoprecipitation , Phosphopyruvate Hydratase/metabolism , Quinoxalines/pharmacology , Rats , Rats, Sprague-Dawley , Time Factors , Transfection
3.
J Endocrinol ; 189(2): 409-21, 2006 May.
Article in English | MEDLINE | ID: mdl-16648306

ABSTRACT

Activin type II receptors (ActRIIs) including ActRIIA and ActRIIB are serine/threonine kinase receptors that form complexes with type I receptors to transmit intracellular signaling of activins, nodal, myostatin and a subset of bone morphogenetic proteins. ActRIIs are unique among serine/threonine kinase receptors in that they associate with proteins having PSD-95, Discs large and ZO-1 (PDZ) domains. In our previous studies, we reported specific interactions of ActRIIs with two independent PDZ proteins named activin receptor-interacting proteins 1 and 2 (ARIP1 and ARIP2). Overexpression of both ARIP1 and ARIP2 reduce activin-induced transcription. Here, we report the isolation of two isoforms of ARIP2 named ARIP2b and 2c. ARIP2, ARIP2b and ARIP2c recognize COOH-terminal residues of ActRIIA that match a PDZ-binding consensus motif. ARIP2 and its isoforms have one PDZ domain in the NH2-terminal region, and interact with ActRIIA. Although PDZ domains containing GLGF motifs of ARIP2b and 2c are identical to that of ARIP2, their COOH-terminal sequences differ from that of ARIP2. Interestingly, unlike ARIP2, overexpression of ARIP2b or 2c did not affect ActRIIA internalization. ARIP2b/2c inhibit inhibitory actions of ARIP2 on activin signaling. ARIP2 is widely distributed in mouse tissues. ARIP2b/2c is expressed in more restricted tissues such as heart, brain, kidneys and liver. Our results indicate that although both ARIP2 and ARIP2b/2c interact with activin receptors, they regulate ActRIIA function in a different manner.


Subject(s)
Activins/metabolism , Adaptor Proteins, Signal Transducing/analysis , Membrane Proteins/analysis , Signal Transduction/physiology , Activin Receptors, Type II/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , CHO Cells , Calcium-Binding Proteins/metabolism , Cells, Cultured , Cricetinae , Endocytosis/physiology , Follicle Stimulating Hormone/metabolism , Isomerism , Kidney/metabolism , Liver/metabolism , Mice , Molecular Sequence Data , Myocardium/metabolism
4.
J Mol Endocrinol ; 36(3): 569-79, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16720724

ABSTRACT

The cytoplasmic immunophilin FKBP12, a 12 kDa FK506-binding protein, has been shown to act as an inhibitor for transforming growth factor-beta (TGF-beta) signaling. FKBP12 binds to the glycine- and serine-rich motif (GS motif) of the TGF-beta type I receptor, and functions as a secure switch to prevent the leaky signal. Upon stimulation with ligand, FKBP12 is released from the receptor to fully propagate the signal. We found that activin, a member of TGF-beta superfamily, also induced the dissociation of FKBP12 from the activin type I receptor (ALK4). However, we observed that the released FKBP12 associates again with the receptor a few hours later. FKBP12 also interacted with another inhibitory molecule of activin signal, Smad7, in an activin-dependent manner, and formed a complex with Smad7 on the type I receptor. FK506, a chemical ligand for FKBP12, which dissociates FKBP12 from the receptor, decreased the interaction between Smad7 and Smad ubiquitin regulatory factor 1 (Smurf1). FK506 also inhibited the ubiquitination of the type I receptor by Smurf1. These findings indicate a new inhibitory function of FKBP12 as an adaptor molecule for the Smad7-Smurf1 complex to regulate the duration of the activin signal.


Subject(s)
Activin Receptors, Type I/metabolism , Smad7 Protein/metabolism , Tacrolimus Binding Protein 1A/metabolism , Ubiquitin-Protein Ligases/metabolism , Activin Receptors, Type I/genetics , Animals , CHO Cells , Cell Line , Cricetinae , Gene Expression Regulation , Humans , Multiprotein Complexes , Signal Transduction/physiology , Smad7 Protein/genetics , Tacrolimus Binding Protein 1A/genetics , Transcription, Genetic , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
5.
J Endocrinol ; 184(3): 493-504, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749808

ABSTRACT

Activin has previously been shown to act as a nerve cell survival factor and to have neurotrophic effects on neurons. However, the role of activin in regulating neurotransmitter expression in the central nervous system and the exact mechanisms involved in this process are poorly understood. In the present study, we report that activin A and basic fibroblast growth factor (bFGF) synergistically increased the protein level of tyrosine hydroxylase (TH), and also greatly increased the TH mRNA level, in both mouse E14 striatal primary cell cultures and the hippocampal neuronal cell line HT22. Activin A and bFGF cooperatively stimulated nuclear translocation of Smad3 and specifically activated ERK1/2, but not p38 or JNK. Interestingly, a specific inhibitor for MEK, U0126, efficiently blocked the induction of TH promoter activity by activin A and bFGF, indicating that activin A collaborated with bFGF signaling to induce the TH gene through selective activation of ERK-type MAP kinase in mouse striatal and HT22 cells. These data suggest that activin A may act in concert with bFGF for the development of TH-positive neurons.


Subject(s)
Activins/pharmacology , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 2/pharmacology , Inhibin-beta Subunits/pharmacology , MAP Kinase Signaling System , Neurons/metabolism , Trans-Activators/metabolism , Tyrosine 3-Monooxygenase/metabolism , Animals , Blotting, Western/methods , Cell Line , Cells, Cultured , Drug Synergism , Immunohistochemistry/methods , Mice , Neurons/drug effects , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Smad3 Protein , Stimulation, Chemical , Tyrosine 3-Monooxygenase/analysis , Tyrosine 3-Monooxygenase/genetics
6.
Jpn J Pharmacol ; 87(2): 134-42, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11700012

ABSTRACT

The protective effect of a brief episode of ischemic preconditioning was examined at an early phase of ischemic-reperfusion injury in the rat kidney. Rats were subjected to 50 min of left renal artery occlusion followed by 120 min of reperfusion. Ischemic preconditioned rats were subjected to preconditioning with two cycles of 3-min ischemia and 5-min reperfusion (IPC). Ischemic-reperfusion injury led to a low recovery of the glomerular filtration rate (GFR). Overt morphological changes, consisting of blood trapping and tubular collapse, were seen. IPC improved the recovery of GFR and renal morphology. The IPC effect was not blocked by 8-(p-sulfophenyl)-theophylline (SPT), a non-selective adenosine receptor antagonist, by 1,3-dipropyl-8-cyclopentylxanthine (DPCPX), a selective A1-receptor antagonist, or by 3,7-dimethyl-1-propargylxanthine (DMPX), a selective A2-receptor antagonist. Intravenous infusion of adenosine (30 microg/min per rat, for 5 min) prior to the 50-min occlusion improved the recovery of GFR, and this protection of GFR was blocked by SPT. Thus, both IPC and exogenous adenosine attenuated ischemic-reperfusion injury of the kidney. However, because three adenosine receptor antagonists failed to abolish the protective effect of IPC, there is no evidence to indicate that activation of adenosine receptors contributes to the IPC effect in the kidney.


Subject(s)
Adenosine/pharmacology , Ischemia/physiopathology , Ischemic Preconditioning , Kidney/blood supply , Receptors, Purinergic P1/physiology , Theobromine/analogs & derivatives , Theophylline/analogs & derivatives , Animals , Blood Pressure/drug effects , Glomerular Filtration Rate/drug effects , Heart Rate/drug effects , Ischemia/etiology , Kidney/physiopathology , Male , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Rats, Wistar , Renal Artery Obstruction/complications , Renal Circulation/drug effects , Reperfusion Injury/etiology , Reperfusion Injury/physiopathology , Theobromine/pharmacology , Theophylline/pharmacology , Time Factors , Ultrasonography, Doppler, Pulsed , Xanthines/pharmacology
7.
Proc Natl Acad Sci U S A ; 98(24): 14022-6, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11698647

ABSTRACT

When neural cells were collected from the entire cerebral cortex of developing mouse fetuses (15.5-17.5 days postcoitum) and their nuclei were transferred into enucleated oocytes, 5.5% of the reconstructed oocytes developed into normal offspring. This success rate was the highest among all previous mouse cloning experiments that used somatic cells. Forty-four percent of live embryos at 10.5 days postcoitum were morphologically normal when premature and early-postmitotic neural cells from the ventricular side of the cortex were used. In contrast, the majority (95%) of embryos were morphologically abnormal (including structural abnormalities in the neural tube) when postmitotic-differentiated neurons from the pial side of the cortex were used for cloning. Whereas 4.3% of embryos cloned with ventricular-side cells developed into healthy offspring, only 0.5% of those cloned with differentiated neurons in the pial side did so. These facts seem to suggest that the nuclei of neural cells in advanced stages of differentiation had lost their developmental totipotency. The underlying mechanism for this developmental limitation could be somatic DNA rearrangements in differentiating neural cells.


Subject(s)
Cerebral Cortex/cytology , Cloning, Organism/methods , Neurons/physiology , Animals , Cell Nucleus/physiology , Cell Transplantation/methods , Cerebral Cortex/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Oocytes
8.
Mol Cell Endocrinol ; 180(1-2): 25-31, 2001 Jun 30.
Article in English | MEDLINE | ID: mdl-11451568

ABSTRACT

Activin signal transduction is regulated through multiple mechanisms. We have identified novel regulatory proteins that control activin functions either intracellularly or extracellularly. As intracellular molecules, PSD-95/Dlg/ZO-1 (PDZ) proteins that specifically associate with activin type II receptors (ActRIIs) were identified. We have named the molecules as activin receptor-interacting proteins (ARIPs). ARIP1 has two WW domains and five PDZ domains, associates not only with ActRIIs but also with Smads, and controls activin functions intracellularly in neuronal cells. Another ARIP we have found has only one PDZ domain, and is likely to be involved in intracellular trafficking and sorting of activin receptor complexes in the cell. As an extracellular regulatory protein, we have identified a novel follistatin-like protein, named follistatin-related gene (FLRG). Like follistatins, FLRG binds activins and bone morphogenetic proteins (BMPs) and controls their functions extracellularly. The mode of association of follistatin and FLRG with activins and their expression patterns are different, suggesting the distinct functions of follistatin and FLRG in vivo.


Subject(s)
Activin Receptors, Type II , Activins/physiology , Proteins , Signal Transduction/drug effects , Activin Receptors/metabolism , Activin Receptors/pharmacology , Activins/antagonists & inhibitors , Activins/metabolism , Activins/pharmacology , Adaptor Proteins, Signal Transducing , Animals , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/physiology , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Follistatin , Follistatin-Related Proteins , Glycoproteins/metabolism , Glycoproteins/pharmacology , Guanylate Kinases , Mice , Protein Binding , Protein Structure, Tertiary , Rats
9.
J Cell Physiol ; 188(2): 236-42, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11424090

ABSTRACT

Bone tissues reportedly contain considerable amounts of activin A and follistatin, an activin A-binding protein. In the present study, we found that follistatin strongly inhibited osteoclast formation in cocultures of mouse bone marrow cells and primary osteoblasts induced by 1alpha,25 dihydroxyvitamin D(3), prostaglandin E(2), and interleukin-1alpha. Antibody aganist activin A also inhibited the osteoclast formation. Furthermore, activin A synergistically stimulated osteoclast differentiation mediated by receptor activator NF-kappaB ligand (RANKL). RT-PCR analysis revealed that osteoblasts produced not only activin A but also follistatin. Western blot analysis of a panel of phosphorylated proteins revealed that activin A stimulated the phosphorylation of p44/42 mitogen activated protein (MAP) kinase (ERK1/2) and p38 MAP kinase in macrophage colony-stimulating factor-dependent bone marrow macrophages (M-BMMPhis). In addition, phosphorylation of Smad2 was observed in M-BMMPhis stimulated with activin A. These findings indicate that the phosphorylation of p44/42 MAP kinase, p38 MAP kinase, and Smad2 is involved in activin A-enhanced osteoclast differentiation induced by RANKL. Taken together, these results suggest that both activin A and follistatin produced by osteoblasts may play an important role in osteoclast differentiation through MAP kinases and Smad2 signaling pathways.


Subject(s)
DNA-Binding Proteins/metabolism , Inhibins/pharmacology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Trans-Activators/metabolism , Activin Receptors , Activins , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/enzymology , Carrier Proteins/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Female , Follistatin , Gene Expression/physiology , Glycoproteins/pharmacology , Growth Substances/pharmacology , Membrane Glycoproteins/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Phosphorylation , RANK Ligand , RNA, Messenger/analysis , Receptor Activator of Nuclear Factor-kappa B , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Smad2 Protein , p38 Mitogen-Activated Protein Kinases
10.
Genomics ; 72(3): 321-30, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11401448

ABSTRACT

Cadherin-related neuronal receptor (CNR) family proteins are known as synaptic cadherins and Reelin receptors. Here we have identified two novel mouse CNR genes, CNRc1 and CNRc2, orthologues of human protocadherin (Pcdh) alpha-c1 and Pcdhalpha-c2, respectively. While the variable large exons of CNRc1 and c2 contain six conserved extracellular cadherin repeats (EC1-6) and are linked to the constant exons, both contain several molecular features distinct from CNR1-8. CNRc1 and c2 lack the Arg-Gly-Asp (RGD) sequence that is conserved in the EC1 of CNR1-8, which is necessary for binding to Reelin. The present studies confirm that CNRc1 and c2 failed to immunoprecipitate with Reelin. In addition, the regulation of novel CNR expression patterns during brain development is slightly different from that of CNR1. The identification of these new CNR genes characterized by their distinct extracellular function and expression is indicative of the novel diversity of the processes of brain structuring and synapse regulation.


Subject(s)
Cadherins/genetics , Multigene Family/genetics , Neuropeptides/genetics , Receptors, Cell Surface , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Brain/embryology , Brain/metabolism , Cadherins/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Line , Cloning, Molecular , DNA/chemistry , DNA/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins , Neuropeptides/metabolism , Phylogeny , Precipitin Tests , Protein Binding , Protocadherins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reelin Protein , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Serine Endopeptidases
11.
Hypertension ; 37(6): 1394-8, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11408383

ABSTRACT

To determine whether angiotensin type 2 (AT(2)) receptor stimulation induces apoptosis in cardiomyocytes in vivo, we developed transgenic mice overexpressing the AT(2) receptor in a cardiac-specific manner, using the alpha-myosin heavy-chain promoter. Ten- to 12-week-old male homozygous transgenic mice (n=44) and wild-type mice (n=44) were used. Both transgenic and wild-type mice were given either saline (control), a subpressor dose of angiotensin II (100 ng. kg(-1). min(-1)), a pressor dose of angiotensin II (1000 ng. kg(-1). min(-1)) for 14 days, a pressor dose of angiotensin II for 28 days to investigate the effects of stimulation on both angiotensin type 1 (AT(1)) and AT(2) receptors, the AT(1) antagonist L158809 alone, or a combination of angiotensin II (1000 ng. kg(-1). min(-1)) and L158809 for 14 days to investigate the effects of selective AT(2) receptor stimulation. Apoptosis was analyzed in paraffin-embedded ventricular sections by the terminal deoxynucleotidyl-transferase-mediated dUTP nick-end labeling (TUNEL) technique. In both transgenic and wild-type mice, administration of a subpressor dose of angiotensin II, L158809, or a combination of angiotensin II and L158809 did not significantly affect the tail-cuff blood pressure or heart-to-body weight ratio, whereas administration of a pressor dose of angiotensin II for 14 or 28 days significantly increased blood pressure and the heart-to-body weight ratio. However, there was no statistical difference between the effects of angiotensin II in transgenic and wild-type mice. The number of TUNEL-positive nuclei was approximately 0 to 10 per 100 000 cardiomyocytes, with no difference between transgenic and wild-type mice, regardless of saline infusion or any stimulation. In infarcted canine myocardial tissue sections for positive control, the number of TUNEL-positive nuclei was increased by 13.8 to 19.1 times compared with those in the noninfarcted myocardium. In conclusion, angiotensin II infusion for a period of 28 days failed to induce cardiomyocyte apoptosis regardless of the presence or absence of cardiac AT(2) receptor overexpression. It is unlikely that in mice the AT(2) receptor is a strong signal to induce cardiomyocyte apoptosis in vivo.


Subject(s)
Apoptosis , Myocardium/cytology , Receptors, Angiotensin/physiology , Angiotensin II/pharmacology , Angiotensin Receptor Antagonists , Animals , Blood Pressure , Body Weight , Heart Rate , Imidazoles/pharmacology , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Organ Size , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Receptors, Angiotensin/genetics , Tetrazoles/pharmacology , Vasoconstrictor Agents/pharmacology
12.
Genes Cells ; 6(2): 151-64, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11260260

ABSTRACT

BACKGROUND: Cadherin-related neuronal receptor (CNR) family genes have been identified in the nervous system by screening molecules bound to Fyn-tyrosine kinase. The CNR family is comprised of diverse synaptic cadherins. The genomic organization of the CNR genes, composed of variable and constant regions, is similar to that of the immunoglobulin gene cluster. The nervous system is characterized by the acquisition of diverse function. This feature is similar to the immune system. In the immune system, the generation and selection of immunoglobulin gene mutants is the underlying basis for acquired immunity. We therefore examined somatic regulation of the CNR family genes in the nervous system to determine whether a similar mechanism controls nervous system development. RESULTS: We first demonstrated that approximately 10% of the CNR3 transcripts were trans-transcripts between the variable and constant exons at the adult stage. Furthermore, somatic mutations of CNR3 transcripts accumulated during brain development, with a marked bias for A-to-G and U-to-C transitions. Approximately 70% of the CNR3 transcripts exhibited a mutation characterized by the addition of a CU-repeat; these transcripts contained seven continuous CU-repeats in the 3' untranslated region at the adult stage, whereas the CNR genomic DNA contained six continuous CU-repeats. Interestingly, a high rate of replacement mutations was detected in the first cadherin domain that functions in acquisition of specificity for cell adhesion in cadherins, while in the other regions the occurrence of silent mutations increased during development. CONCLUSION: The present report is the first description of the generation and possible selection of somatic mutations of synaptic cadherin transcripts. The somatic alterations of CNR family transcripts and their synaptic localization have interesting implications for the molecular basis underlying the establishment of somatic rearrangement of neuronal networks.


Subject(s)
Cadherins/genetics , Mutation , Nervous System/metabolism , RNA, Messenger/genetics , Synapses/metabolism , Amino Acid Sequence , Animals , Base Sequence , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Nervous System/embryology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
13.
Jpn J Pharmacol ; 85(1): 109-13, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11243564

ABSTRACT

We investigated whether a compensatory vasoconstrictor action would be induced by a hypotensive intervention in the hindquarter vascular region of conscious spontaneously hypertensive rats (SHRs). Mean arterial pressure and hindquarter blood flow were recorded. After hemorrhage (withdrawing blood, 0.3 ml/100 g body weight), hindquarter resistance (HQR) was increased significantly. The decrease in HQR induced by the administration of a ganglionic blocker (C6; 25 mg/kg, i.v.) was significantly greater in SHRs with hemorrhage than in those without hemorrhage. The present results suggest that a detectable hindquarter compensator tone occurs due to hemorrhage in SHRs, although an abnormal substantial vasoconstrictor tone already exists in the hindquarters.


Subject(s)
Hemorrhage/physiopathology , Hindlimb/blood supply , Vascular Resistance/physiology , Vasoconstriction/physiology , Animals , Aorta, Abdominal , Blood Flow Velocity , Blood Pressure/physiology , Consciousness , Ganglionic Blockers/pharmacology , Hexamethonium/pharmacology , Hypotension/physiopathology , Male , Rats , Rats, Inbred SHR , Rats, Wistar , Regional Blood Flow/physiology , Vascular Resistance/drug effects
14.
Cell Signal ; 12(8): 565-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-11027950

ABSTRACT

We demonstrate the difference between the follistatin isoforms (FS-288 and FS-315), two activin-binding proteins, in the neutralizing activity for activin signalling. Transcriptional reporter assay using 3TP-Lux, an activin-responsive reporter construct, showed that the inhibitory effect of FS-288 on activin-induced transcriptional response is more potent than that of FS-315. The potency was not influenced by the presence of heparan sulfates, by which FS, in particular FS-288, associates with cell surfaces at a high affinity. Furthermore, FS-288 inhibited the binding of activin to its type II receptor more markedly than did FS-315, as evidenced by surface plasmon resonance and affinity cross-linking experiments. Moreover, the Kd of FS-288 and FS-315 for activin A was estimated to be 46.5+/-0.37 pM and 432+/-26 pM, respectively, by surface plasmon resonance experiments. These results indicate that the different potency between the two FS isoforms in the inhibition of activin activities depends on their affinity for activin A.


Subject(s)
Glycoproteins/metabolism , Inhibins/antagonists & inhibitors , Signal Transduction , Activins , Biosensing Techniques , Follistatin , Humans , Inhibins/metabolism , Inhibins/physiology , K562 Cells , Macromolecular Substances , Protein Isoforms/metabolism , Surface Plasmon Resonance , Transcriptional Activation
15.
J Biol Chem ; 275(52): 40788-96, 2000 Dec 29.
Article in English | MEDLINE | ID: mdl-11010968

ABSTRACT

Follistatin is an activin-binding protein that prevents activin from binding to its receptors and neutralizes its activity. Follistatin also binds bone morphogenetic proteins (BMPs). In this study, we report the identification of a novel follistatin-like protein from mouse. The mouse cDNA encodes a 256-residue precursor and most likely a mouse homologue of human FLRG, which was found at the breakpoint of the chromosomal rearrangement in a B-cell line. Whereas follistatin has three follistatin domains, which are presumed to be growth factor binding motifs, FLRG possesses only two follistatin domains. Northern blotting revealed that mRNAs for FLRG were abundantly expressed in heart, lung, kidney, and testis in mouse. The recombinant mouse FLRG proteins were found to have binding activity for both activin and bone morphogenetic protein-2. Like follistatin, FLRG has higher affinity for activin than for BMP-2. The FLRG protein inhibited activin-induced and BMP-2-induced transcriptional responses in a dose-dependent manner. Glutathione S-transferase fusion proteins encoding various regions of FLRG were produced and studied. Ligand blotting using (125)I-activin revealed that the COOH-terminal region containing the second follistatin domain was able to bind activin. Our finding implies that cellular signaling by activin and BMPs is tightly regulated by multiple members of the follistatin family.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Carrier Proteins/isolation & purification , Inhibins/metabolism , Proteins/isolation & purification , Transforming Growth Factor beta , Activins , Animals , Binding Sites , Bone Morphogenetic Protein 2 , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins/chemistry , Carrier Proteins/physiology , Cloning, Molecular , Dose-Response Relationship, Drug , Follistatin-Related Proteins , HeLa Cells , Humans , Mice , Proteins/chemistry , Proteins/physiology , RNA, Messenger/analysis , Recombinant Proteins , Transcription, Genetic/drug effects
16.
Biochemistry ; 39(21): 6390-400, 2000 May 30.
Article in English | MEDLINE | ID: mdl-10828953

ABSTRACT

Oocytes of the starfish, Asterina pectinifera, are arrested at the G2 phase of meiosis I and possess a prominent germinal vesicle in which maternal stores of nuclear proteins which are destined for use primarily by the early embryo are stored. Germinal vesicle breakdown and subsequent oocyte maturation is triggered by activation of the p34(cdc2)/cyclin B complex, which is present as the preform in the cytoplasm. The aim of the present study was to identify and biochemically characterize in vivo substrates of the kinase. Two nucleic acid binding nuclear proteins designated NAAP1 and NAAP2 were found, both of which contain 345 amino acid residues with pI 3. 6 and which serve as substrates. The only difference between the two proteins was in the primary amino acid sequence at position 51, which is Asn in NAAP1 but Thr in NAAP2. NAAPs are phosphorylated in vivo during oocyte maturation but not at the meiotic G(2) stage. NAAPs are phosphorylated in vitro by the cdc2 kinase on the same site as in vivo. Although there are other evolutionarily conserved consensus sequences for phosphorylation by mitotically active cdc2 kinase in NAAPs and NAAP-derived fragments containing the sequences were efficiently phosphorylated in vitro, these sites in the intact NAAPs were not phosphorylated either in vivo or in vitro. These results suggest that the tertiary structure of NAAPs affects the target specificity of the cdc2 kinase.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Oocytes/physiology , Amino Acid Sequence , Animals , CDC2 Protein Kinase/metabolism , Consensus Sequence , Conserved Sequence , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/isolation & purification , DNA-Binding Proteins/metabolism , Evolution, Molecular , Female , G2 Phase , Humans , Meiosis , Molecular Sequence Data , Nuclear Proteins/isolation & purification , Oocytes/cytology , Peptide Fragments/chemistry , Phosphorylation , Sea Urchins , Sequence Alignment , Sequence Homology, Amino Acid , Starfish , Xenopus laevis
17.
Int J Dev Biol ; 44(3): 327-30, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10853830

ABSTRACT

We compared the expression patterns of follistatin and two follistatin-related proteins (FRP and m7365) during early mouse development. m7365 is expressed continuously during preimplantation development, in contrast to FRP and follistatin. At early postimplantation stages, follistatin and 7365 are expressed from E6.0, while FRP is detected from E7.5 onwards. Although there is some overlap between the expression of these genes in the primitive streak and somites, their overall expression patterns are distinct.


Subject(s)
Blastocyst/metabolism , Embryo, Mammalian/metabolism , Glycoproteins/biosynthesis , Animals , DNA, Complementary/metabolism , Follistatin , Follistatin-Related Proteins , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Distribution
18.
J Endocrinol Invest ; 23(4): 231-4, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10853708

ABSTRACT

Activin A is a dimeric glycoprotein showing a high sequence homology with transforming growth factor-beta (TGF-beta) and playing autocrine/paracrine actions in reproductive tissues. However, since the synthesis of activin is ubiquitous it may have a role in regulating cell growth and differentiation in several tissues. Previous studies showed that activin A is expressed by insulin-positive B cells of human pancreatic islets, and women with gestational diabetes have higher serum activin A levels than healthy pregnant women at the same gestational age. The present study aimed to evaluate the effect of activin A on insulin secretion from cultured human pancreatic islets. With this purpose human pancreatic islets were incubated with varying concentrations of activin A (0.1 to 10.0 nM). In absence of glucose, activin A did not modify insulin secretion at the different concentrations used. In absence of activin A, 8.3 mM and 16.7 mM glucose significantly increased insulin secretion, with a dose-dependent pattern. In presence of a non stimulatory concentration of glucose (3.3 mM), activin A significantly increased insulin secretion starting from low concentration (0.1 nM). Furthermore, the addition of activin A to 8.3 mM and 16.7 mM glucose induced an additional effect of the dose-dependent glucose-mediated insulin secretion (p<0.001). The present data could support a role for activin A in human endocrine pancreas in modulating insulin response to different glucose concentrations.


Subject(s)
Inhibins/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Activins , Culture Techniques , Dose-Response Relationship, Drug , Glucose/administration & dosage , Glucose/pharmacology , Humans , Inhibins/administration & dosage , Insulin Secretion
19.
Cancer Lett ; 155(1): 37-46, 2000 Jul 03.
Article in English | MEDLINE | ID: mdl-10814877

ABSTRACT

TSC-36 (TGF-beta1-stimulated clone 36) is a TGF-beta1 inducible gene whose product is an extracellular glycoprotein that contains a single follistatin module. TSC-36 is highly expressed in the lung, but its physiological function is unknown. In an attempt to elucidate it, we investigated the effect of TSC-36 on proliferation of human lung cancer cell lines. We found a correlation between expression of TSC-36 and cell growth: TSC-36 mRNA was not detected in cells derived from small cell lung cancer (SCLC) cells, a highly aggressive neoplasm, but was detected in some non-small cell lung cancer (NSCLC) cells, a moderately aggressive neoplasm. This suggested an antiproliferative function for TSC-36. To address this question, NSCLC PC-14 cells, which express very low level of TSC-36 protein, were transfected with TSC-36 cDNA and the proliferative capacity of stable transfectants was determined by measuring the doubling time, colony forming activity in soft agar and the level of incorporation of (3)H-thymidine into DNA. Under normal culture conditions, the transfected cells showed a longer doubling time, lower plating efficiency and lower rate of DNA synthesis than the parental cells and the control neo transfectant cells. These findings suggested that expression of TSC-36 caused growth inhibition in human lung cancer cells.


Subject(s)
Glycoproteins/biosynthesis , Glycoproteins/genetics , Lung Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Blotting, Northern , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Small Cell/metabolism , Cell Division/drug effects , Flow Cytometry , Follistatin-Related Proteins , Humans , Plasmids , RNA, Messenger/metabolism , Transfection , Tumor Cells, Cultured
20.
Eur J Endocrinol ; 142(5): 506-11, 2000 May.
Article in English | MEDLINE | ID: mdl-10802530

ABSTRACT

OBJECTIVE: To evaluate the effect of the growth factor activin A on the secretion of prolactin (PRL) and GH in cultured GH3 cells. METHODS: The concentrations of PRL and GH secreted from GH3 cells cultured in media with and without activin A were measured by RIA, and the expression of PRL mRNA and GH mRNA were analyzed using the Northern blot method. RESULTS: Activin A significantly inhibited PRL release from GH3 cells cultured for 48h in a dose-dependent manner (activin: 0.3-3nM). The inhibitory effects of 3nM activin A were observed in the culture from 12h to 48h (53.2% of control). Activin A (3nM) also significantly inhibited the expression of PRL mRNA at 24h (33.8% of control). In contrast, activin A significantly stimulated GH release from GH3 cells cultured for 48h in a dose-dependent manner (activin: 0.3-3nM). The stimulatory effect of 3nM activin A was observed in the culture for 48h (157.6% of control). Activin A (3nM) also significantly stimulated the expression of GH mRNA at 24h (183.6% of control). In spite of these significant changes in PRL and GH secretion, pit-1 mRNA levels were not significantly changed by activin A. CONCLUSIONS: These findings indicated that activin A modulates PRL and GH secretion through the regulation of PRL and GH gene transcription in GH3 cells, but that these effects are unrelated to pit-1 gene expression.


Subject(s)
Growth Hormone/biosynthesis , Growth Hormone/metabolism , Inhibins/metabolism , Pituitary Neoplasms/metabolism , Prolactin/biosynthesis , Prolactin/metabolism , Activins , Animals , Blotting, Northern , Gene Expression , Growth Hormone/genetics , Prolactin/genetics , RNA, Messenger/analysis , Radioimmunoassay , Rats , Time Factors , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...