Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Control Release ; 188: 67-77, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-24892974

ABSTRACT

The ubiquitin-proteasome system is central in the regulation of cellular proteins controlling cell cycle progression and apoptosis, drawing much interest for developing effective targeted cancer therapies. Herein, we developed a novel pH-responsive polymeric-micelle-based carrier system to effectively deliver the proteasome inhibitor MG132 into cancer cells. MG132 is covalently bound to the block copolymer composed of polyethylene glycol (PEG) and polyaspartate through an acid-labile hydrazone bond. This bond is stable at physiological condition, but hydrolytically degradable in acidic compartments in the cell, such as late-endosomes and lysosomes, and thus, it was used for controlled release of MG132 after EPR-mediated preferential accumulation of the micelles into the tumor. MG132-loaded micelles have monodispersed size distribution with an average diameter of 45nm, and critical micelle concentration is well below 10(-7)M. In vitro studies against several cancer cell lines confirmed that MG132-loaded micelles retained the cytotoxic effect, and this activity was indeed due to the inhibition of proteasome by released MG132 from the micelles. Real-time in vitro confocal-microscopy experiments clearly indicated that MG132-conjugated micelles disintegrated only inside the target cells. By intravital confocal micro-videography, we also confirmed the prolonged circulation of MG132 loaded micelles in the bloodstream, which lead to tumor specific accumulation of micelles, as confirmed by in vivo imaging 24h after injection. These micelles showed significantly lower in vivo toxicity than free MG132, while achieving remarkable antitumor effect against a subcutaneous HeLa-luc tumor model. Our findings create a paradigm for future development of polymeric-micelle-based carrier system for other peptide aldehyde type proteasome inhibitors to make them effective cohort of the existing cancer therapeutic regiments.


Subject(s)
Antineoplastic Agents/administration & dosage , Delayed-Action Preparations/chemistry , Leupeptins/administration & dosage , Micelles , Proteasome Inhibitors/administration & dosage , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Humans , Hydrogen-Ion Concentration , Leupeptins/pharmacokinetics , Leupeptins/therapeutic use , Mice, Inbred BALB C , Neoplasms/drug therapy , Neoplasms/pathology , Polymers/chemistry , Proteasome Inhibitors/pharmacokinetics , Proteasome Inhibitors/therapeutic use
2.
Gene Ther ; 18(9): 910-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21451578

ABSTRACT

Duchenne muscular dystrophy (DMD) is the most common form of the progressive muscular dystrophies characterized by defects of the dystrophin gene. Although primarily characterized by degeneration of the limb muscles, cardiomyopathy is a major cause of death. Therefore, the development of curative modalities such as gene therapy is imperative. We evaluated the cardiomyopathic features of mdx mice to observe improvements in response to intravenous administration of recombinant adeno-associated virus (AAV) type 9 encoding microdystrophin. The myocardium was extensively transduced with microdystrophin to significantly prevent the development of fibrosis, and expression persisted for the duration of the study. Intraventricular conduction patterns, such as the QRS complex duration and S/R ratio in electrocardiography, were also corrected, indicating that the transduced microdystrophin has a protective effect on the dystrophin-deficient myocardium. Furthermore, BNP and ANP levels were reduced to normal, suggesting the absence of cardiac dysfunction. In aged mice, prevention of ectopic beats as well as echocardiographic amelioration was also demonstrated with improved exercise performance. These findings indicate that AAV-mediated cardiac transduction with microdystrophin might be a promising therapeutic strategy for the treatment of dystrophin-deficient cardiomyopathy.


Subject(s)
Cardiomyopathies/prevention & control , Dependovirus/genetics , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Aging , Animals , Dystrophin/chemistry , Fibrosis , Genetic Vectors , Humans , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/complications , Myocardium/pathology , Transduction, Genetic , Transgenes
3.
Gene Ther ; 16(3): 392-403, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19037241

ABSTRACT

We previously reported the development of a prototype 'oncolytic Sendai virus (SeV) vector' formed by introducing two major genomic modifications to the original SeV, namely deletion of the matrix (M) gene to avoid budding of secondary viral particles and manipulation of the trypsin-dependent cleavage site of the fusion (F) gene to generate protease-specific sequences. As a result, the 'oncolytic SeV' that was susceptible to matrix metalloproteinases (MMPs) was shown to selectively kill MMP-expressing tumors through syncytium formation in vitro and in vivo. However, its efficacy has been relatively limited because of the requirement of higher expression of MMPs and smaller populations of MMP-expressing tumors. To overcome these limitations, we have designed an optimized and dramatically powerful oncolytic SeV vector. Truncation of 14-amino acid residues of the cytoplasmic domain of F protein resulted in dramatic enhancement of cell-killing activities of oncolytic SeV, and the combination with replacement of the trypsin cleavage site with the new urokinase type plasminogen activator (uPA)-sensitive sequence (SGRS) led a variety of human tumors, including prostate (PC-3), renal (CAKI-I), pancreatic (BxPC3) and lung (PC14) cancers, to extensive death through massive cell-to-cell spreading without significant dissemination to the surrounding noncancerous tissue in vivo. These results indicate a dramatic improvement of antitumor activity; therefore, extensive utility of the newly designed uPA-targeted oncolytic SeV has significant potential for treating patients bearing urokinase-expressing cancers in clinical settings.


Subject(s)
Genetic Vectors , Neoplasms/therapy , Oncolytic Virotherapy/methods , Sendai virus/genetics , Urokinase-Type Plasminogen Activator/genetics , Animals , Female , Giant Cells/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mutagenesis, Site-Directed , Neoplasm Transplantation , Neoplasms/enzymology , Neoplasms/pathology , Oncolytic Viruses/genetics , Peptide Fragments/genetics , Transplantation, Heterologous , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/metabolism
4.
Gene Ther ; 11(14): 1137-45, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15085175

ABSTRACT

Malignant tumor cells often express matrix metalloproteinases (MMPs) at a high level to enable their dissemination and metastasis. Sendai virus (SeV), a nonsegmented negative strand RNA virus, spreads in the target tissues in vivo via cleavage activation of the viral fusion glycoprotein by a tissue-specific, trypsin-like enzyme. By deleting the viral matrix protein, we previously generated a recombinant SeV that does not bud to mature virions, but is highly fusogenic and spreads extensively from cell to cell in a trypsin-dependent manner. Here, we changed the tryptic cleavage site of the fusion glycoprotein of this virus to a site susceptible to MMPs. The resulting recombinant virus was no longer activated by trypsin but spread efficiently in cultured cells supplemented with MMP2 or MMP9 and in human tumor cell lines expressing these MMPs. Furthermore, the virus spread extensively in tumor cells xenotrasplanted to nude mice without disseminating to the surrounding normal cells, leading to the inhibition of the tumor growth in the mice. These results demonstrate the selective targeting and killing of human tumor cells by recombinant SeV technology and greatly advance the reemerging concept of oncolytic virotherapy, which currently appears to rely largely upon a natural preference of certain viruses for cancer cells.


Subject(s)
Genetic Therapy/methods , Matrix Metalloproteinases/metabolism , Neoplasms/therapy , Recombinant Fusion Proteins/administration & dosage , Sendai virus/genetics , Animals , Cell Line, Tumor , Genetic Engineering , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Mice , Mice, Nude , Neoplasms/virology , Recombinant Fusion Proteins/metabolism , Transfection/methods
5.
Cell Growth Differ ; 12(11): 573-80, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11714638

ABSTRACT

Expression of membrane-type (MT) 5 matrix metalloproteinase (MMP) in the mouse brain was examined. MT5-MMP was expressed in the cerebrum in embryos, but it declined after birth. In contrast, expression in the cerebellum started to increase postnatally and continued thereafter. The cells expressing MT5-MMP were postmitotic neurons that showed gelatinolytic activities. Specific expression of MT5-MMP was observed in the neurons but not in the glial cells when embryonal mouse carcinoma P19 cells were differentiated in vitro by retinoic acid treatment. Neurons isolated from dorsal root ganglia also expressed MT5-MMP, and it was localized at the edge of growth cone. Proteoglycans inhibit neurite extension and regulate synaptogenesis. The inhibitory effect of the proteoglycans on neurite extension of dorsal root ganglia neurons was effectively eliminated by recombinant MT5-MMP. Thus, MT5-MMP expressed in neurons may play a role in axonal growth that contributes to the regulation of neural network formation.


Subject(s)
Cell Differentiation , Metalloendopeptidases/metabolism , Neurons/cytology , Neurons/enzymology , Animals , Base Sequence , Blotting, Northern , Brain/cytology , Brain/embryology , Brain/enzymology , Brain/growth & development , Cell Differentiation/drug effects , Cell Size/drug effects , Cloning, Molecular , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Growth Cones/drug effects , In Situ Hybridization , Matrix Metalloproteinases, Membrane-Associated , Mice , Molecular Sequence Data , Neurites/drug effects , Neurites/enzymology , Neurons/drug effects , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tretinoin/pharmacology , Tumor Cells, Cultured
6.
J Cell Biol ; 153(5): 893-904, 2001 May 28.
Article in English | MEDLINE | ID: mdl-11381077

ABSTRACT

Migratory cells including invasive tumor cells frequently express CD44, a major receptor for hyaluronan and membrane-type 1 matrix metalloproteinase (MT1-MMP) that degrades extracellular matrix at the pericellular region. In this study, we demonstrate that MT1-MMP acts as a processing enzyme for CD44H, releasing it into the medium as a soluble 70-kD fragment. Furthermore, this processing event stimulates cell motility; however, expression of either CD44H or MT1-MMP alone did not stimulate cell motility. Coexpression of MT1-MMP and mutant CD44H lacking the MT1-MMP-processing site did not result in shedding and did not promote cell migration, suggesting that the processing of CD44H by MT1-MMP is critical in the migratory stimulation. Moreover, expression of the mutant CD44H inhibited the cell migration promoted by CD44H and MT1-MMP in a dominant-negative manner. The pancreatic tumor cell line, MIA PaCa-2, was found to shed the 70-kD CD44H fragment in a MT1-MMP-dependent manner. Expression of the mutant CD44H in the cells as well as MMP inhibitor treatment effectively inhibited the migration, suggesting that MIA PaCa-2 cells indeed use the CD44H and MT1-MMP as migratory devices. These findings revealed a novel interaction of the two molecules that have each been implicated in tumor cell migration and invasion.


Subject(s)
Cell Movement , Hyaluronan Receptors/metabolism , Leucine/analogs & derivatives , Metalloendopeptidases/metabolism , Phenylalanine/analogs & derivatives , Amino Acid Sequence , Animals , Cell Movement/drug effects , Cell Size/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/enzymology , Extracellular Matrix/metabolism , Genes, Dominant/genetics , Humans , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/genetics , Leucine/pharmacology , Ligands , Matrix Metalloproteinase 14 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenylalanine/pharmacology , Plant Proteins/pharmacology , Protein Processing, Post-Translational/drug effects , Sequence Deletion/genetics , Solubility , Sulfones/pharmacology , Thiophenes/pharmacology , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Trypsin Inhibitors , Tumor Cells, Cultured , alpha-Amylases/antagonists & inhibitors
7.
J Biol Chem ; 274(48): 34260-6, 1999 Nov 26.
Article in English | MEDLINE | ID: mdl-10567400

ABSTRACT

Among the five membrane-type matrix metalloproteinases (MT-MMPs), MT1-, MT2-, MT3-, and MT5-MMPs have about a 20-amino acid cytoplasmic tail following the transmembrane domain. In contrast, a putative transmembrane domain of MT4-MMP locates at the very C-terminal end, and the expected cytoplasmic tail is very short or nonexistent. Such sequences often act as a glycosylphosphatidylinositol (GPI) anchoring signal rather than as a transmembrane domain. We thus examined the possibility that MT4-MMP is a GPI-anchored proteinase. Our results showed that [(3)H]ethanolamine, which can be incorporated into the GPI unit, specifically labeled the MT4-MMP C-terminal end in a sequence-dependent manner. In addition, phosphatidylinositol-specific phospholipase C treatment released the MT4-MMP from the surface of transfected cells. These results indicate that MT4-MMP is the first GPI-anchored proteinase in the MMP family. During cultivation of the transfected cells, MT4-MMP appeared to be shed from the cell surface by the action of an endogenous metalloproteinase. GPI anchoring of MT4-MMP on the cell surface indicates a unique biological function and character for this proteinase.


Subject(s)
Glycosylphosphatidylinositols/metabolism , Matrix Metalloproteinases/metabolism , Metalloendopeptidases , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , COS Cells , Chlorocebus aethiops , Cricetinae , Cricetulus , Endopeptidases/drug effects , Endopeptidases/metabolism , Ethanolamine/metabolism , Fluorescent Antibody Technique, Indirect , Humans , Matrix Metalloproteinases/chemistry , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases, Membrane-Associated , Mice , Microscopy, Confocal , Molecular Sequence Data , Phenylalanine/analogs & derivatives , Phenylalanine/pharmacology , Phosphatidylinositol Diacylglycerol-Lyase , Phosphoinositide Phospholipase C , Protease Inhibitors/pharmacology , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Sequence Alignment , Thiophenes/pharmacology , Tritium , Type C Phospholipases/pharmacology
8.
FEBS Lett ; 457(3): 353-6, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10471807

ABSTRACT

Five distinct membrane-type matrix metalloproteinases (MT-MMP) have been reported by cDNA cloning. However, the mt4-mmp gene product (MMP-17) has not been identified yet in spite of the cDNA isolation [Puente et al. (1996), Cancer Res. 56, 944-949]. In this study, we re-examined the transcripts for human mt4-mmp by 5' RACE and identified two types of transcripts. The minor one corresponded to the cDNA reported by Puente et al. and failed to express protein, and the other is the major transcript that has an extended open reading frame and expressed 67 and 71 kDa translation products. Thus, functional mt4-mmp has been identified for the first time.


Subject(s)
DNA, Complementary/isolation & purification , Matrix Metalloproteinases , Metalloendopeptidases/genetics , Amino Acid Sequence , Animals , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , COS Cells , Carcinoma/enzymology , Carcinoma/genetics , DNA Primers , Humans , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/immunology , Metalloendopeptidases/metabolism , Mice , Molecular Sequence Data , Nucleic Acid Amplification Techniques , Protein Biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Tumor Cells, Cultured
10.
Gene ; 211(1): 57-62, 1998 Apr 28.
Article in English | MEDLINE | ID: mdl-9573338

ABSTRACT

Three genes potentially encoding novel matrix metalloproteinases (MMPs) were identified by sequence similarity searching of Caenorhabditis elegans genome database, and cDNAs for these MMPs were cloned. The predicted gene products (MMP-C31,-H19 and -Y19) display a similar domain organization to human MMPs. MMP-H19 and -Y19 are unique in that they have an RXKR motif between the propeptide and catalytic domains that is a furin-like cleavage site, and conserved only in stromelysin-3 and membrane-type MMPs. The amino acid sequence homology with MMP-1/human interstitial collagenase at the catalytic domain is 45%, 34% and 23% for MMP-C31, -H19 and -Y19, respectively. Recombinant proteins of C. elegans MMPs cleaved an MMP peptide substrate with efficiency proportional to their amino acid homology with human MMPs. Digestion of gelatin was observed only with MMP-C31. Enzyme activity of MMP-C31 and -H19 was inhibited by human tissue inhibitor of MMPs (TIMP)-1, TIMP-2 and synthetic MMP inhibitors, BB94 and CT543, indicating that the catalytic sites of these C. elegans MMPs are structurally closely related with those of mammalian MMPs.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans/enzymology , Metalloendopeptidases/chemistry , Animals , Cloning, Molecular , Databases, Factual , Enzyme Inhibitors/pharmacology , Escherichia coli/genetics , Gelatin/metabolism , Genes, Helminth/genetics , Helminth Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Analysis , Sequence Homology, Amino Acid , Substrate Specificity
11.
Biochemistry ; 36(23): 7225-38, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9188724

ABSTRACT

The hatching enzyme (envelysin) of the sea urchin Hemicentrotus pulcherrimus was purified from the medium of hatched blastulae. By cDNA cloning its deduced amino acid sequence and molecular architecture were revealed. The 591-residue precursor with calculated Mr of 66,123 consists of an 18-residue signal sequence, a 151-residue propeptide, and a 422-residue mature enzyme with N-terminal catalytic and C-terminal hemopexin-like domains. As compared with that of Paracentrotus lividus, its amino acid sequence is 69% identical and 10% similar. They share typical structural features with the mammalian MMP gene family members: cysteine switch, zinc-binding signature, methionine-turn, Cys residues near both ends of hemopexin-like domain, etc. However, its propeptide has a 70-residue extra sequence with an Asp- and Glu-rich stretch, supposedly involved in the proenzyme activation by binding Ca2+ ions in seawater. The hinge region is also longer than those of most MMPs, with an extra sequence rich in Thr and Arg residues. Mature 50K enzyme is highly susceptible to autolytic cleavage at Gln(503)-Leu(504), producing the 38K form retaining catalytic activity and substrate specificity against fertilization envelope. The 38K form and 15K fragment were coeluted from a gel-filtration column, suggesting that these two fragments are disulfide-bridged and that the tertiary structure is not much deviated. The 38K form further autolyzed to 32K form by cleaving Tyr(450)-Tyr(451) bond with the loss of protein-substrate specificity, retaining only nonspecific protease activity. Thus, the autolytic release of 2/3 of the C-terminal domain reduced the highly specific enzyme to a common nonspecific protease, implying that the size and structure of almost the entire hemopexin-like domain is essential for the protein substrate specificity. Moreover, autolytic degradation of envelysins from the two species follow quite different pathways despite their high homology in structure. The 38K and 32K forms were inhibited by bovine TIMP-1 with different IC50 values, indicating that its inhibitory activity depends on the extent of the interaction with the C-terminal domain of the enzyme.


Subject(s)
Embryo, Nonmammalian/enzymology , Metalloendopeptidases/genetics , Amino Acid Sequence , Animals , Autolysis , Base Sequence , Cattle , Chromatography, High Pressure Liquid , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Glycoproteins/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/isolation & purification , Metalloendopeptidases/metabolism , Molecular Sequence Data , Molecular Weight , Peptide Mapping , Protease Inhibitors/pharmacology , Sea Urchins , Sequence Alignment , Substrate Specificity , Tissue Inhibitor of Metalloproteinases
12.
Cancer Res ; 56(24): 5678-83, 1996 Dec 15.
Article in English | MEDLINE | ID: mdl-8971175

ABSTRACT

Membrane-type matrix metalloproteinase 1 (MT1-MMP) is a member of the recently identified unique membrane-type subgroup in the matrix metalloproteinase (MMP) family. MT1-MMP has proteolytic activity against components in the extracellular matrix and activates progelatinase A (72-kDa type IV procollagenase/proMMP-2) on the cell surface. Because MT1-MMP is frequently expressed in a variety of tumors, we examined its contribution to their metastatic potential. The mouse lung carcinoma cell line Madison 109 was transiently transfected with a MT1-MMP expression plasmid and inoculated into the tail vein of BALB/c mouse. Fate of the transfected cells was monitored by the neo(r) gene in the plasmid using the quantitative PCR method. The survival rate of the parental cells in lung was 0.7% of the inoculated cells. It was increased by 3-fold with the MT1-MMP transfected cells and the number of the lung nodules increased accordingly. Immunostaining of the consecutive tissue sections revealed that lung nodules expressing MT1-MMP were positive for gelatinase A as well, whereas MT1-MMP-negative cells were not stained for gelatinase A at all. Thus, MT1-MMP-expressing cells acquire specific ability to bind exogenous progelatinase A.


Subject(s)
Gelatinases/metabolism , Lung Neoplasms/secondary , Metalloendopeptidases/metabolism , Neoplasm Proteins/metabolism , Animals , Antisense Elements (Genetics)/genetics , Cell Adhesion , Fibrosarcoma/enzymology , Gelatinases/genetics , Gene Expression , Genetic Vectors , Humans , Matrix Metalloproteinase 14 , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/genetics , Mice , Neoplasm Proteins/genetics , Transfection , Tumor Cells, Cultured
13.
J Cell Sci ; 109 ( Pt 5): 953-9, 1996 May.
Article in English | MEDLINE | ID: mdl-8743942

ABSTRACT

Matrix metalloproteinases (MMPs), which degrade the components of the extracellular matrix, are key enzymes involved in the tissue remodeling of multicellular organisms. Since MMPs are secreted as inactive zymogens (pro-MMPs), they have to be activated to function. We identified a membrane-type MMP (MT-MMP) that activated proMMP-2 (pro-gelatinase A = 72 kDa type IV pro-collagenase) and described its expression on the invasive tumor cell surface. In this study we further examined the expression and role of MT-MMP in the activation of proMMP-2 during mouse embryogenesis. Northern blotting demonstrated that MT-MMP expression was increased together with that of MMP-2 and its inhibitor gene, TIMP-2, in embryos depending upon the number of days after gestation, and decreased with maturation after birth. In situ hybridization and immunohistochemistry localized MT-MMP mRNA and protein in the cells of ossifying tissues where both MMP-2 and TIMP-2 were expressed. Activated MMP-2 was detected by gelatin zymography in the lysates prepared from the micro dissected tissues that expressed the three genes. The activation rate of proMMP-2 was proportional to the expression of MMP-2 and MT-MMP. These results indicated that proMMP-2 activation through its activator, MT-MMP, is a physiological system used by organisms to initiate tissue remodeling on the cell surface.


Subject(s)
Enzyme Precursors/metabolism , Gelatinases/analysis , Metalloendopeptidases/biosynthesis , Protease Inhibitors/pharmacology , Proteins/analysis , Amino Acid Sequence , Animals , Embryonic and Fetal Development/physiology , Gelatinases/antagonists & inhibitors , Immunohistochemistry , In Situ Hybridization , Matrix Metalloproteinase 2 , Matrix Metalloproteinases, Membrane-Associated , Metalloendopeptidases/analysis , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Molecular Sequence Data , RNA, Messenger/biosynthesis , Substrate Specificity , Tissue Inhibitor of Metalloproteinase-2
14.
Zoolog Sci ; 11(1): 153-6, 1994 Feb.
Article in English | MEDLINE | ID: mdl-7765182

ABSTRACT

A cDNA with 2.3 kb encoding F1-F0 ATP synthase (proton ATPase) beta-subunit homologue was isolated from a testis cDNA library of the sea urchin, Hemicentrotus pulcherrimus. The deduced amino acid sequence consisted of 523 residues which contained a 19-residue amino-terminal signal peptide and a 8-residue glycine-rich consensus sequences. Analysis of poly(A) +RNA and/or total RNA from H. pulcherrimus testis, ovary, unfertilized eggs, and embryos by Northern blot revealed a 2.4 kb RNA.


Subject(s)
Proton-Translocating ATPases/genetics , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary , Molecular Sequence Data , Proton-Translocating ATPases/chemistry , Sea Urchins , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...