Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
Add more filters










Publication year range
1.
FASEB J ; 15(6): 988-94, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11292659

ABSTRACT

Tumor growth and metastasis are critically dependent on the formation of new blood vessels. The present study found that extracellular matrix protein 1 (ECM1), a newly described secretory glycoprotein, promotes angiogenesis. This was initially suggested by in situ hybridization studies of mouse embryos indicating that the ECM1 message was associated with blood vessels and its expression pattern was similar to that of flk-1, a recognized marker for endothelium. More direct evidence for the role of ECM1 in angiogenesis was provided by the fact that highly purified recombinant ECM1 stimulated the proliferation of cultured endothelial cells and promoted blood vessel formation in the chorioallantoic membrane of chicken embryos. Immunohistochemical staining with specific antibodies indicated that ECM1 was expressed by the human breast cancer cell lines MDA-435 and LCC15, both of which are highly tumorigenic. In addition, staining of tissue sections from patients with breast cancer revealed that ECM1 was present in a significant proportion of primary and secondary tumors. Collectively, the results of this study suggest that ECM1 possesses angiogenic properties that may promote tumor progression.


Subject(s)
Angiogenesis Inducing Agents/physiology , Extracellular Matrix Proteins/physiology , Neovascularization, Physiologic/physiology , Angiogenesis Inducing Agents/biosynthesis , Breast Neoplasms , Cell Division/physiology , Disease Progression , Endothelium/cytology , Extracellular Matrix Proteins/biosynthesis , Humans , Tumor Cells, Cultured , Up-Regulation
2.
Bone ; 28(1): 14-20, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11165938

ABSTRACT

Two mRNAs are transcribed from the extracellular matrix protein 1 gene (Ecm1): Ecm1a and an alternatively spliced Ecm1b. We studied Ecm1 mRNA expression and localization during endochondral bone formation and investigated the effect of recombinant human (rh) Ecm1a protein on organ cultures of embryonic mouse metatarsals. Of the two transcripts, Ecm1a mRNA was predominantly expressed in fetal metacarpals from day 16 to 19 after gestation. Ecm1 expression was not found in 16- and 17-day-old metatarsals of which the perichondrium was removed. In situ hybridization and immunohistochemistry demonstrated Ecm1 expression in the connective tissues surrounding the developing bones, but not in the cartilage. Biological effects of rhEcm1a protein on fetal metatarsal cultures were biphasic: at low concentrations, Ecm1a stimulated alkaline phosphatase activity and had no effect on mineralization, whereas at higher concentrations, Ecm1a dose dependently inhibited alkaline phosphatase activity and mineralization. These results suggest that Ecm1a acts as a novel negative regulator of endochondral bone formation.


Subject(s)
Alkaline Phosphatase/metabolism , Calcification, Physiologic/physiology , Extracellular Matrix Proteins/metabolism , Metatarsal Bones/metabolism , Animals , Calcium Radioisotopes , Cell Differentiation/physiology , Chondrocytes/chemistry , Chondrocytes/cytology , Chondrocytes/enzymology , Extracellular Matrix Proteins/analysis , Extracellular Matrix Proteins/genetics , Forelimb/embryology , Gene Expression Regulation, Developmental/physiology , Humans , Metatarsal Bones/cytology , Metatarsal Bones/embryology , Mice , Paracrine Communication/physiology , RNA, Messenger/analysis , Recombinant Proteins/metabolism
3.
J Biol Chem ; 276(7): 4909-16, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11060285

ABSTRACT

MPIF-1, a CC chemokine, is a specific inhibitor of myeloid progenitor cells and is the most potent activator of monocytes. The solution structure of myeloid progenitor inhibitor factor-1 (MPIF-1) has been determined by NMR spectroscopy. The structure reveals that MPIF-1 is a monomer with a well defined core except for termini residues and adopts the chemokine fold of three beta-strands and an overlying alpha-helix. In addition to the four cysteines that characterize most chemokines, MPIF-1 has two additional cysteines that form a disulfide bond. The backbone dynamics indicate that the disulfide bonds and the adjacent residues that include the functionally important N-terminal and N-terminal loop residues show significant dynamics. MPIF-1 is a highly basic protein (pI >9), and the structure reveals distinct positively charged pockets that could be correlated to proteoglycan binding. MPIF-1 is processed from a longer proprotein at the N terminus and the latter is also functional though with reduced potency, and both proteins exist as monomers under a variety of solution conditions. MPIF-1 is therefore unique because longer proproteins of all other chemokines oligomerize in solution. The MPIF-1 structure should serve as a template for future functional studies that could lead to therapeutics for preventing chemotherapy-associated myelotoxicity.


Subject(s)
Chemokines, CC/chemistry , Amino Acid Sequence , Animals , Chemokines, CC/genetics , Chemokines, CC/metabolism , Escherichia coli/genetics , Heparin/metabolism , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Structure, Secondary , Sequence Homology, Amino Acid , Transformation, Bacterial , Ultracentrifugation
4.
Mol Cell Endocrinol ; 162(1-2): 131-44, 2000 Apr 25.
Article in English | MEDLINE | ID: mdl-10854706

ABSTRACT

Stanniocalcin (STC) is a polypeptide hormone that was first discovered in fish and recently identified in humans and other mammals. In fish STC is produced by one gland, circulates freely in the blood and plays an integral role in mineral homeostasis. In mammals, STC is produced in a number of different tissues and serves a variety of different functions. In kidney, STC regulates phosphate reabsorption by proximal tubule cells, whereas in ovary it appears to be involved in steroid hormone synthesis. However there is no information on circulating levels of STC in mammals or the regulation of its secretion. In this report we have developed a radioimmunoassay (RIA) for human STC. The RIA was validated for measuring tissue hormone levels. However human and other mammalian sera were completely devoid of immunoreactive STC (irSTC). To explore the possibility that mammalian STC might have a short half-life pharmacokinetic analysis was carried out in rats. STC pharmacokinetics were best described by a two compartment model where the distribution phase (t1/2(alpha)) equaled 1 min and the elimination phase (t1/2(beta)) was 60 min. However the STC in the elimination phase no longer crossreacted in the RIA indicating it had undergone substantial chemical modification, which could explain our inability to detect irSTC in mammalian sera. When we compared the pharmacokinetics of human and fish STC in mammalian and fish models the human hormone was always eliminated faster, indicating that human STC has unique structural properties. There also appears to be a unique clearance mechanism for STC in mammals. Hence there are major differences in the delivery and biology of mammalian STC. Unlike fishes, mammalian STC does not normally circulate in the blood and functions instead as a local mediator of cell function. Future studies will no doubt show that this has had important ramifications on function as well.


Subject(s)
Glycoproteins/analysis , Hormones/analysis , Radioimmunoassay/methods , Animals , Cattle , Female , Glycoproteins/blood , Glycoproteins/metabolism , Half-Life , Hormones/blood , Hormones/metabolism , Humans , Immunohistochemistry , Kinetics , Male , Models, Biological , Oncorhynchus mykiss , Rats , Rats, Wistar , Recombinant Proteins/analysis , Recombinant Proteins/blood , Recombinant Proteins/pharmacokinetics , Tissue Distribution
5.
Nucleic Acids Res ; 28(3): 809-17, 2000 Feb 01.
Article in English | MEDLINE | ID: mdl-10637334

ABSTRACT

The yeast CCR4-NOT protein complex is a global regulator of RNA polymerase II transcription. It is comprised of yeast NOT1 to NOT5, yeast CCR4 and additional proteins like yeast CAF1. Here we report the isolation of cDNAs encoding human NOT2, NOT3, NOT4 and a CAF1-like factor, CALIF. Analysis of their mRNA levels in different human tissues reveals a common ubiquitous expression pattern. A multitude of two-hybrid interactions among the human cDNAs suggest that their encoded proteins also form a complex in mammalian cells. Functional conservation of these proteins throughout evolution is supported by the observation that the isolated human NOT3 and NOT4 cDNAs can partially com-plement corresponding not mutations in yeast. Interestingly, human CALIF is highly homologous to, although clearly different from, a recently described human CAF1 protein. Conserved interactions of this factor with both NOT and CCR4 proteins and co-immunoprecipitation experiments suggest that CALIF is a bona fide component of the human CCR4-NOT complex.


Subject(s)
Cell Cycle Proteins/metabolism , Conserved Sequence/genetics , Fungal Proteins/metabolism , Proteins , Repressor Proteins/metabolism , Ribonucleases , Saccharomyces cerevisiae Proteins , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Cell Line , Cloning, Molecular , Expressed Sequence Tags , Fungal Proteins/genetics , Gene Expression Profiling , Genes, Fungal/genetics , Genetic Complementation Test , Humans , Molecular Sequence Data , Mutation/genetics , Precipitin Tests , Protein Binding , RNA, Messenger/analysis , RNA, Messenger/genetics , Receptors, CCR4 , Receptors, Chemokine , Repressor Proteins/chemistry , Repressor Proteins/genetics , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Two-Hybrid System Techniques
6.
Ann N Y Acad Sci ; 923: 25-42, 2000.
Article in English | MEDLINE | ID: mdl-11193762

ABSTRACT

Rabbit uteroglobin is the founder member of a family of mammalian proteins that has expanded to more than 20 members within the last few years. All members are small, secretory, rarely glycosylated dimeric proteins with unclear physiological functions and are mainly expressed in mucosal tissues. A phylogenetic analysis shows that the family can be grouped into five subfamilies, A to E. Subfamily A contains rabbit uteroglobin and its orthologues from various species; most of these have been described to form antiparallel homodimers via two intermolecular disulfide bonds. All other subfamily members contain a third conserved cysteine and, from existing biochemical data, it can be predicted that a member of subfamily B or C will likely form heterodimers with a partner from subfamily E or D, respectively. Besides the mentioned cysteines, only one central lysine is conserved in all family members. In the known uteroglobin structures, this lysine forms an exposed salt bridge with an aspartate side chain, which is conserved in almost all sequences. Using radiation hybrid mapping and P1 clone analysis and utilizing data from the human genome project, we show that all known five human family members (Clara cell 10-kDa protein, lipophilins A and B, lacryglobin, mammaglobin) and a new member, we call lymphoglobin, are localized on chromosome 11q12.2 in a dense cluster spanning not more than approximately 400 kbp.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Genitalia, Female/metabolism , Globins/genetics , Multigene Family/physiology , Sequence Homology , Uteroglobin/genetics , Amino Acid Sequence/genetics , Animals , Female , Genitalia, Female/cytology , Humans , Mammaglobin A , Molecular Sequence Data , Myelin Proteins/genetics , Neoplasm Proteins/genetics , Phylogeny , Proteolipids/genetics , Radiation Hybrid Mapping , Secretoglobins , Uteroglobin/chemistry
7.
Oncogene ; 18(47): 6496-504, 1999 Nov 11.
Article in English | MEDLINE | ID: mdl-10597252

ABSTRACT

Recently a new member of the human tumor necrosis factor (TNF) family named as VEGI was reported. However, very little is known about the biological activities displayed by this cytokine. In this report, we show that in myeloid cells VEGI activated the transcription factor kappa B (NF-kappa B) as determined by the electrophoretic mobility shift assay, induced degradation of I kappa B alpha, and nuclear translocation of p65 subunit of NF-kappa B. VEGI also activated NF-kappa B-dependent reporter gene expression. In addition, VEGI activated c-Jun N-terminal kinase. When examined for growth modulatory effects, VEGI inhibited the proliferation of breast carcinoma (MCF-7), epithelial (HeLa), and myeloid (U-937 and ML-1a) tumor cells; and activated caspase-3 leading to PARP cleavage. VEGI-induced cytotoxicity was potentiated by inhibitors of protein synthesis. VEGI also induced proliferation of normal human foreskin fibroblast cells. The activity of VEGI could neither be neutralized by antibodies against TNF, nor could it compete with TNF binding, indicating that the activity of VEGI is not due to TNF and it binds to a distinct receptor. These results suggest that VEGI, a new member of the TNF family, has a signaling pathway similar to TNF and is most likely a multifunctional cytokine.


Subject(s)
Cell Division/physiology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/physiology , Base Sequence , DNA Primers , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases , Protein Binding , Recombinant Proteins/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15 , Tumor Necrosis Factor-alpha/metabolism
8.
Cell Biol Int ; 23(1): 81-8, 1999.
Article in English | MEDLINE | ID: mdl-10527550

ABSTRACT

The protein encoded by the proto-oncogene c-fos is constitutively nuclear in most cell types analyzed. It has a predicted molecular weight of about 55 kDa. Proteins with a molecular weight above 40 kDa cannot enter the nucleus passively. Our interest was to study which regions in the protein are involved in the nuclear transport. We prepared a series of deletions and point mutations of the protein and cloned the mutated genes into a eukaryotic expression vector. Cos-1 cells were used to express the mutants transiently. Using indirect immunofluorescence we studied the subcellular localization, analyzing the percentage of cells containing the protein in the nucleus, the cytoplasm, or both locations. Our studies showed that the Fos protein contains several regions which can act independently to translocate the protein into the nucleus.


Subject(s)
Cell Nucleus/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Biological Transport , COS Cells , Fluorescent Antibody Technique, Indirect , Point Mutation , Proto-Oncogene Proteins c-fos/genetics
9.
Science ; 285(5425): 260-3, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10398604

ABSTRACT

The tumor necrosis factor (TNF) superfamily of cytokines includes both soluble and membrane-bound proteins that regulate immune responses. A member of the human TNF family, BLyS (B lymphocyte stimulator), was identified that induced B cell proliferation and immunoglobulin secretion. BLyS expression on human monocytes could be up-regulated by interferon-gamma. Soluble BLyS functioned as a potent B cell growth factor in costimulation assays. Administration of soluble recombinant BLyS to mice disrupted splenic B and T cell zones and resulted in elevated serum immunoglobulin concentrations. The B cell tropism of BLyS is consistent with its receptor expression on B-lineage cells. The biological profile of BLyS suggests it is involved in monocyte-driven B cell activation.


Subject(s)
B-Lymphocytes/immunology , Lymphocyte Activation , Membrane Proteins/physiology , Monocytes/immunology , Tumor Necrosis Factor-alpha/physiology , Amino Acid Sequence , Animals , B-Cell Activating Factor , B-Cell Activation Factor Receptor , B-Lymphocyte Subsets/immunology , Cell Line , Cells, Cultured , Humans , Immunoglobulins/blood , Interferon-gamma/pharmacology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Receptors, Cytokine/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Recombinant Proteins/pharmacology , Sequence Alignment , Species Specificity , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation
10.
Proc Natl Acad Sci U S A ; 96(7): 3700-5, 1999 Mar 30.
Article in English | MEDLINE | ID: mdl-10097100

ABSTRACT

A serpin was identified in normal mammary gland by differential cDNA sequencing. In situ hybridization has detected this serpin exclusively in the myoepithelial cells on the normal and noninvasive mammary epithelial side of the basement membrane and thus was named myoepithelium-derived serine proteinase inhibitor (MEPI). No MEPI expression was detected in the malignant breast carcinomas. MEPI encodes a 405-aa precursor, including an 18-residue secretion signal with a calculated molecular mass of 46 kDa. The predicted sequence of the new protein shares 33% sequence identity and 58% sequence similarity to plasminogen activator inhibitor (PAI)-1 and PAI-2. To determine whether MEPI can modulate the in vivo growth and progression of human breast cancers, we transfected a full-length MEPI cDNA into human breast cancer cells and studied the orthotopic growth of MEPI-transfected vs. control clones in the mammary fat pad of athymic nude mice. Overexpression of MEPI inhibited the invasion of the cells in the in vitro invasion assay. When injected orthotopically into nude mice, the primary tumor volumes, axillary lymph node metastasis, and lung metastasis were significantly inhibited in MEPI-transfected clones as compared with controls. The expression of MEPI in myoepithelial cells may prevent breast cancer malignant progression leading to metastasis.


Subject(s)
Breast Neoplasms/pathology , Neoplasm Proteins , Serpins/physiology , Adult , Amino Acid Sequence , Animals , Cell Division , Female , Humans , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Open Reading Frames , Organ Specificity , Plasminogen Activator Inhibitor 1/chemistry , Plasminogen Activator Inhibitor 2/chemistry , Pregnancy , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serpins/chemistry , Serpins/genetics , Transcription, Genetic , Transfection , Tumor Cells, Cultured
11.
FASEB J ; 13(1): 181-9, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9872942

ABSTRACT

A novel member of the tumor necrosis factor (TNF) family has been identified from the human umbilical vein endothelial cell cDNA library, named vascular endothelial growth inhibitor (VEGI). The VEGI gene was mapped to human chromosome 9q32. The cDNA for VEGI encodes a protein of 174 amino acid residues with the characteristics of a type II transmembrane protein. Its amino acid sequence is 20-30% identical to other members of the TNF family. Unlike other members of the TNF family, VEGI is expressed predominantly in endothelial cells. Local production of a secreted form of VEGI via gene transfer caused complete suppression of the growth of MC-38 murine colon cancers in syngeneic C57BL/6 mice. Histological examination showed marked reduction of vascularization in MC-38 tumors that expressed soluble but not membrane-bound VEGI or were transfected with control vector. The conditioned media from soluble VEGI-expressing cells showed marked inhibitory effect on in vitro proliferation of adult bovine aortic endothelial cells. Our data suggest that VEGI is a novel angiogenesis inhibitor of the TNF family and functions in part by directly inhibiting endothelial cell proliferation. The results further suggest that VEGI maybe highly valuable toward angiogenesis-based cancer therapy.


Subject(s)
Antineoplastic Agents/metabolism , Colonic Neoplasms/physiopathology , Neovascularization, Pathologic , Tumor Necrosis Factor-alpha/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Division , Cell Line , DNA, Complementary , Endothelium, Vascular/cytology , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Solubility , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 15 , Tumor Necrosis Factor-alpha/genetics
12.
Anal Biochem ; 267(1): 125-34, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9918664

ABSTRACT

Peptide mapping and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) were conducted to characterize the human genome-based recombinant proteins. Accurate mass values for the deleted forms of the myeloid progenitor inhibitory factor chemokine (MPIF-1d23), and the keratinocyte growth factor (KGF-2d33) were measured as 8848.55 +/- 0.25 and 16,175.87 +/- 0.89 Da, respectively. The mass accuracy of delayed ion extraction MALDI-MS measurements was within 20 ppm of the cDNA predicted value. Reduction and alkylation of the chemokine showed the presence of six cysteine residues and three disulfide bonds. Additional confirmation of disulfide bonding among the cysteine residues of the chemokine was demonstrated by identifying the RP-HPLC separated tryptic and endoprotease Glu-C peptides. Three methionine residues of the chemokine were identified by MALDI-MS of its cyanogen bromide (CNBr) cleavage products. The KGF-2d33 growth factor, however, lacked the disulfide bonding between the two-cysteine residues and contained two free sulfhydryl groups. Direct analysis of the growth factor CNBr digest showed 7542.99, 4993.4, and 3107.7 Da peptides, which identified the methionine residues. Peptide mapping mass spectrometry indicated that host-specific posttranslational modifications had not influenced the gene expression work.


Subject(s)
Chemokines, CC/chemistry , Fibroblast Growth Factors , Growth Substances/chemistry , Peptide Mapping/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Amino Acid Sequence , Chemokines, CC/genetics , Chromatography, High Pressure Liquid , Disulfides/chemistry , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Growth Substances/genetics , Humans , Molecular Sequence Data , Molecular Weight , Peptide Fragments/chemistry , Peptide Fragments/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
13.
Cytotechnology ; 29(3): 215-20, 1999 May.
Article in English | MEDLINE | ID: mdl-19003344

ABSTRACT

A recombinant Chinese hamster ovary clone was cultivated in a 2L Cytopilot Mini fluidized bed bioreactor using Cytoline 1 microcarriers and a 10L B. Braun stirred tank bioreactor with Cytodex 1 microcarriers. Cytoline 1 is a macroporous polyethylene microcarrier and Cytodex 1 is a solid DEAE-dextran microcarrier. Cytoline 1 microcarriers in the fluidized bed bioreactor were gently mixed by an uplifting flow. Circulation and sparging in Cytopilot Mini were separated from the fluidized microcarrier bed. Cytopilot Mini bioreactor with Cytoline 1 microcarriers offered 2.3 times more surface area than the stirred tank bioreactor. The 2L fluidized bed bioreactor accommodated approximately half the cells in the 10L stirred tank bioreactor. Moreover, Cytopilot Mini had approximately three times more product output rate and 5.5 times higher specific productivity than the stirred tank bioreactor.

14.
Cytotechnology ; 29(2): 151-8, 1999 Mar.
Article in English | MEDLINE | ID: mdl-22359063

ABSTRACT

A large number of microcarriers are commercially available. The capability of cells to successfully proliferate on microcarriers varies with cell lines and media. Choosing the right microcarrier for a particular cell line is more than a choice of a microcarrier. It is part of an integrated process design. A detailed picture of cell growth and product formation will not only be essential in identifying the kind of microcarrier, but also in determining other parts of the process, such as operation mode and media. Our initial screening on thirteen microcarriers showed that cultures on some microcarriers reached a low cell density but high cell-specific productivity, and high density microcarrier cultures have a low specific productivity. The result is a similar product output per unit volume and time for these two types of cultures. An ideal culture system shall have increased volumetric productivity at elevated cell density. This requires the process goal to be incorporated as early as cell line construction and screening. A high output process can then be realized through high density culture.

15.
Blood ; 92(6): 2123-32, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9731071

ABSTRACT

We describe the characterization of siglec-5 (sialic acid-binding Ig-like lectin-5), a novel transmembrane member of the immunoglobulin superfamily, highly related to the myeloid antigen, CD33. A full-length cDNA encoding siglec-5 was isolated from a human activated monocyte cDNA library. Sequencing predicted that siglec-5 contains four extracellular immunoglobulin-like domains, the N-terminal two of which are 57% identical to the corresponding region of CD33. The cytoplasmic tail is also related to that of CD33, containing two tyrosine residues embodied in immunoreceptor tyrosine-based inhibitory motif-like motifs. The siglec-5 gene was shown to map to chromosome 19q13.41-43, closely linked to the CD33 gene. When siglec-5 was expressed on COS cells or as a recombinant protein fused to the Fc region of human IgG1, it was able to mediate sialic acid-dependent binding to human erythrocytes and soluble glycoconjugates, suggesting that it may be involved in cell-cell interactions. By using specific antibodies, siglec-5 was found to have an expression pattern distinct from that of CD33, being present at relatively high levels on neutrophils but absent from leukemic cell lines representing early stages of myelomonocytic differentiation. Western blot analysis of neutrophil lysates indicated that siglec-5 exists as a disulfide-linked dimer of approximately 140 kD.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, Myelomonocytic/biosynthesis , Membrane Glycoproteins/biosynthesis , Amino Acid Sequence , Antigens, CD/blood , Antigens, CD/genetics , Antigens, Differentiation, Myelomonocytic/blood , Antigens, Differentiation, Myelomonocytic/genetics , Carbohydrate Conformation , Cell Line , Erythrocytes/metabolism , Humans , Lectins/blood , Leukemia, Myeloid/metabolism , Membrane Glycoproteins/blood , Membrane Glycoproteins/genetics , Molecular Sequence Data , N-Acetylneuraminic Acid/blood , Neutrophils/chemistry , Rosette Formation , Sequence Homology, Amino Acid , Sialic Acid Binding Ig-like Lectin 3 , Tumor Cells, Cultured
16.
J Clin Invest ; 102(6): 1142-51, 1998 Sep 15.
Article in English | MEDLINE | ID: mdl-9739048

ABSTRACT

LIGHT is a new member of tumor necrosis factor (TNF) cytokine family derived from an activated T cell cDNA library. LIGHT mRNA is highly expressed in splenocytes, activated PBL, CD8(+) tumor infiltrating lymphocytes, granulocytes, and monocytes but not in the thymus and the tumor cells examined. Introduction of LIGHT cDNA into MDA-MB-231 human breast carcinoma caused complete tumor suppression in vivo. Histological examination showed marked neutrophil infiltration and necrosis in LIGHT expressing but not in the parental or the Neo-transfected MDA-MB-231 tumors. Interferon gamma (IFNgamma) dramatically enhances LIGHT-mediated apoptosis. LIGHT protein triggers apoptosis of various tumor cells expressing both lymphotoxin beta receptor (LTbetaR) and TR2/HVEM receptors, and its cytotoxicity can be blocked specifically by addition of a LTbetaR-Fc or a TR2/HVEM-Fc fusion protein. However, LIGHT was not cytolytic to the tumor cells that express only the LTbetaR or the TR2/HVEM or hematopoietic cells examined that express only the TR2/HVEM, such as PBL, Jurkat cells, or CD8(+) TIL cells. In contrast, treatment of the activated PBL with LIGHT resulted in release of IFNgamma. Our data suggest that LIGHT triggers distinct biological responses based on the expression patterns of its receptors on the target cells. Thus, LIGHT may play a role in the immune modulation and have a potential value in cancer therapy.


Subject(s)
Apoptosis , Genes, Tumor Suppressor , Membrane Proteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Receptors, Virus/metabolism , Tumor Necrosis Factor-alpha/metabolism , Chromosome Mapping , Culture Media, Serum-Free , Female , Gene Transfer Techniques , Humans , In Situ Hybridization, Fluorescence , Interferon-gamma/metabolism , Ligands , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating , Lymphotoxin beta Receptor , Male , Membrane Proteins/genetics , Receptors, Tumor Necrosis Factor, Member 14 , Tumor Cells, Cultured , Tumor Necrosis Factor Ligand Superfamily Member 14 , Tumor Necrosis Factor-alpha/genetics
17.
J Biol Chem ; 273(38): 24797-804, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-9733783

ABSTRACT

A previously undescribed human member of the cystatin superfamily called cystatin F has been identified by expressed sequence tag sequencing in human cDNA libraries. A full-length cDNA clone was obtained from a library made from mRNA of CD34-depleted cord blood cells. The sequence of the cDNA contained an open reading frame encoding a putative 19-residue signal peptide and a mature protein of 126 amino acids with two disulfide bridges and enzyme-binding motifs homologous to those of Family 2 cystatins. Unlike other human cystatins, cystatin F has 2 additional Cys residues, indicating the presence of an extra disulfide bridge stabilizing the N-terminal region of the molecule. Recombinant cystatin F was produced in a baculovirus expression system and characterized. The mature recombinant protein processed by insect cells had an N-terminal segment 7 residues longer than that of cystatin C and displayed reversible inhibition of papain and cathepsin L (Ki = 1.1 and 0.31 nM, respectively), but not cathepsin B. Like cystatin E/M, cystatin F is a glycoprotein, carrying two N-linked carbohydrate chains at positions 36 and 88. An immunoassay for quantification of cystatin F showed that blood contains low levels of the inhibitor (0.9 ng/ml). Six B cell lines in culture secreted barely detectable amounts of cystatin F, but several T cell lines and especially one myeloid cell line secreted significant amounts of the inhibitor. Northern blot analysis revealed that the cystatin F gene is primarily expressed in peripheral blood cells and spleen. Tissue expression clearly different from that of the ubiquitous inhibitor, cystatin C, was also indicated by a high incidence of cystatin F clones in cDNA libraries from dendritic and T cells, but no clones identified by expressed sequence tag sequencing in several B cell libraries and in >600 libraries from other human tissues and cells.


Subject(s)
Cystatins/chemistry , Cystatins/genetics , Amino Acid Sequence , Base Sequence , Biomarkers, Tumor , Cloning, Molecular , Cystatin C , Cystatins/biosynthesis , DNA, Complementary , Databases, Factual , Gene Library , Glycoproteins/chemistry , Glycosylation , Humans , Molecular Sequence Data , Molecular Weight , Phylogeny , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid
18.
FASEB J ; 12(10): 845-54, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9657524

ABSTRACT

A newly identified member of the tumor necrosis factor receptor (TNFR) superfamily shows activities associated with osteoclastogenesis inhibition and fibroblast proliferation. This new member, called TR1, was identified from a search of an expressed sequence tag database, and encodes 401 amino acids with a 21-residue signal sequence. Unlike other members of TNFR, TR1 does not contain a transmembrane domain and is secreted as a 62 kDa glycoprotein. TR1 gene maps to chromosome 8q23-24.1 and its mRNA is abundantly expressed on primary osteoblasts, osteogenic sarcoma cell lines, and primary fibroblasts. The receptors for TR1 were detected on a monocytic cell line (THP-1) and in human fibroblasts. Scatchard analyses indicated two classes of high and medium-high affinity receptors with a kD of approximately 45 and 320 pM, respectively. Recombinant TR1 induced proliferation of human foreskin fibroblasts and potentiated TNF-induced proliferation in these cells. In a coculture system of osteoblasts and bone marrow cells, recombinant TR1 completely inhibited the differentiation of osteoclast-like multinucleated cell formation in the presence of several bone-resorbing factors. TR1 also strongly inhibited bone-resorbing function on dentine slices by mature osteoclasts and decreased 45Ca release in fetal long-bone organ cultures. Anti-TR1 monoclonal antibody promoted the formation of osteoclasts in mouse marrow culture assays. These results indicate that TR1 has broad biological activities in fibroblast growth and in osteoclast differentiation and its functions.


Subject(s)
Bone Resorption/physiopathology , Osteoclasts/physiology , Receptors, Tumor Necrosis Factor/genetics , Animals , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/genetics , Cell Division , Chromosome Mapping , Chromosomes, Human, Pair 8 , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Infant , Male , Mice , Monocytes/metabolism , Osteoclasts/cytology , Osteoprotegerin , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/physiology , Tumor Cells, Cultured
19.
Placenta ; 19(4): 307-14, 1998 May.
Article in English | MEDLINE | ID: mdl-9639327

ABSTRACT

This study investigated whether primary culture of human amniotic membrane cells (PCHAM) could be used as an in vitro model system for the study of interferon (IFN) production. PCHAM cells infected with Newcastle disease virus (NDV) produced the two antigenic types of IFN, previously shown in a amniotic membrane cells (HAM) system. PCHAM IFN was detected as early as 2 h after NDV infection and was composed by two antigenically distinct fractions, one neutralized with anti-HuIFN beta antibody and another that is not related to IFN beta, -alpha and -gamma. These fractions correspond respectively to 80 and 20 per cent of the IFN produced 4 h after virus induction, 55 and 45 per cent of the IFN produced from 4 to 12 h and 67 and 33 per cent of the IFN produced 12 h after virus induction. A cDNA library, established from PCHAM with or without NDV infection, was screened for IFN alpha and -beta using specific primers. The PCR product, amplified by IFN beta primers, was cloned, sequenced and expressed in Escherichia coli M15. The sequences of several cloned cDNAs were identical to HuIFN beta gene and the antiviral activity of the expressed protein was neutralized only by antiHuIFN-beta antibody. The other IFN fraction not neutralized by polyclonal antibodies anti-IFN beta, -alpha and -gamma is now being studied.


Subject(s)
Amnion/cytology , Amnion/metabolism , Interferons/biosynthesis , Amnion/immunology , Base Sequence , Cloning, Molecular , Coculture Techniques , DNA Primers/genetics , Female , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferons/genetics , Kinetics , Polymerase Chain Reaction , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
DNA Cell Biol ; 17(5): 449-60, 1998 May.
Article in English | MEDLINE | ID: mdl-9628588

ABSTRACT

Herein, we describe the analysis and mapping of cDNA clones encoding variant forms of the human homolog of the canine 180-kDa ribosome receptor (p180). One form, similar to the chicken ES/130 homolog, possesses a large uninterrupted C-terminal region composed predominantly of heptad repeats predicted to form an alpha-helical double-stranded coiled-coil rod. Other forms contain in addition a 10-amino acid consensus motif, NQGKKAEGAQ, repeated up to 54 times in tandem close to the N-terminus. Such repeats in canine p180 represent a ribosome-binding domain. The cDNA hybridized to a major 6-kb transcript in all tissues examined, where very high expression was observed in tissues that carry out a high level of secretion such as pancreas, liver, and placenta. The ES130/p180 gene was mapped to chromosome 20p12, and a potential pseudogene appears to reside on chromosome 7. In summary, the data suggest that p180 exists in humans in different forms because of complete removal of tandem repeats, or partial intraexonic splicing, creating different repeat lengths with potentially novel ribosome-binding characteristics.


Subject(s)
Avian Proteins , Receptors, Cytoplasmic and Nuclear/chemistry , Ribosomes/metabolism , Alternative Splicing , Animals , Cell Line , Chickens , Chromosome Mapping , Chromosomes, Human, Pair 20/genetics , Dogs , Endothelium, Vascular , Extracellular Matrix Proteins/chemistry , HL-60 Cells , Humans , Molecular Weight , Protein Structure, Secondary , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/isolation & purification , Repetitive Sequences, Nucleic Acid , Sequence Homology, Amino Acid , Transcription, Genetic , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...