Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Clin Obes ; 7(4): 216-221, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28397375

ABSTRACT

As childhood obesity increases, it is becoming important to understand the complications of obesity in children and develop novel biomarkers. Evidence indicates that microRNAs (miRNA) are dys-regulated in obesity and may serve as sensitive and specific circulating biomarkers. Non-alcoholic fatty liver disease (NAFLD) is a complication of obesity that ultimately requires a liver biopsy to determine disease severity. While studies have been conducted in adults, no study to date has examined circulating miRNAs in children with obesity and NAFLD. The goal of this study was to evaluate a panel of selected circulating miRNAs in obese children compared to healthy controls. We present here an analysis of a pre-selected panel of 20 candidate miRNAs in obese children compared to healthy controls. The miRNAs were chosen based on having been previously reported to be involved in NAFLD. We found that 16 out of 20 miRNAs tested were elevated at least twofold in children with obesity compared to controls. miR-122 and miR-199a showed the greatest increase in children with obesity versus controls. Both also had a high area under the curve when receiver-operator curves were plotted. Several circulating miRNAs correlated with body mass index (BMI) or serum transaminases. This study provides initial evidence that circulating miRNAs can be measured in the paediatric population and provides several diagnostic candidates increased in children with obesity that may be relevant to NAFLD.


Subject(s)
MicroRNAs/blood , Pediatric Obesity/blood , Adolescent , Biomarkers , Case-Control Studies , Child , Female , Humans , Male , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/complications , Pediatric Obesity/complications
2.
Gut ; 55(6): 856-62, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16361307

ABSTRACT

BACKGROUND: Connective tissue growth factor (CCN2) is upregulated in pancreatic fibrosis and desmoplastic pancreatic tumours. CCN2 interacts with integrin alpha5beta1 on pancreatic stellate cells (PSC) in which it stimulates fibrogenesis, adhesion, migration, and proliferation. AIM: To determine the structural domain(s) in CCN2 that interact with integrin alpha5beta1 to regulation PSC functions. METHODS: Primary activated rat PSC were tested for their adherence to isoforms of CCN2 comprising modules 1-4 (CCN2(1-4)), modules 3-4 (CCN2(3-4)), module 3 alone (CCN2(3)), or module 4 alone (CCN2(4)). Adhesion studies were performed in the presence of EDTA, divalent cations, anti-integrin alpha5beta1 antibodies, CCN2 synthetic peptides, or heparin, or after pretreatment of the cells with heparinase, chondroitinase, or sodium chlorate. CCN2 integrin alpha5beta1 binding was analysed in cell free systems. The ability of CCN2(1-4), CCN2(3-4), or CCN2(4) to stimulate PSC migration was evaluated in the presence of anti-integrin alpha5beta1 or heparin. RESULTS: PSC adhesion was stimulated by CCN2(1-4), CCN2(3-4), or CCN2(4) and supported by Mg2+ but not Ca2+. CCN2(4) supported PSC adhesion or migration were blocked by anti-integrin alpha5beta1 antibodies or by treatment of cells with heparinase or sodium chlorate. A direct interaction between CCN2(4) and integrin alpha5beta1 was demonstrated in cell free assays. The sequence GVCTDGR in module 4 mediated the binding between CCN2(4) and integrin alpha5beta1 as well as CCN2(4) mediated PSC adhesion and migration. CONCLUSIONS: A GVCTDGR sequence in module 4 of CCN2 is a novel integrin alpha5beta1 binding site that is essential for CCN2 stimulated functions in PSC and which represents a new therapeutic target in PSC mediated fibrogenesis.


Subject(s)
Immediate-Early Proteins/physiology , Integrin alpha5beta1/metabolism , Intercellular Signaling Peptides and Proteins/physiology , Pancreas/cytology , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Movement/physiology , Cell-Free System , Cells, Cultured , Connective Tissue Growth Factor , Culture Media, Conditioned , Dose-Response Relationship, Drug , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Pancreas/drug effects , Pancreas/physiology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
3.
J Clin Pathol ; 57(4): 422-5, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15047749

ABSTRACT

BACKGROUND: Desmoplastic small round cell tumour (DSRCT) is a rare and often fatal abdominal tumour that is distinguished by well defined islands of cells, surrounded by prominent desmoplastic stroma. As in certain other tumours, the function of the Wilms's tumour protein (WT1) in repressing gene transcription is lost in DSRCT. AIMS: To assess the expression and localisation of connective tissue growth factor (CCN2) in DSRCT because this protein is transcriptionally repressed by WT1 and is associated with the production of abundant extracellular matrix. METHODS: CCN2 was assessed by in situ hybridisation and immunohistochemistry. RESULTS: CCN2 mRNA and protein were colocalised to the tumour cells themselves, in addition to stromal fibroblasts and vascular endothelial cells. CONCLUSIONS: These data show that CCN2 is produced in high amounts by several cell types in DSRCT, and highlight a potential role for this factor in the autocrine and paracrine regulation of tumour cell growth, matrigenesis, and angiogenesis.


Subject(s)
Immediate-Early Proteins/analysis , Intercellular Signaling Peptides and Proteins/analysis , Neoplasm Proteins/analysis , Peritoneal Neoplasms/chemistry , Adolescent , Child , Connective Tissue Growth Factor , Endothelial Cells/chemistry , Endothelium, Vascular/chemistry , Fibroblasts/chemistry , Humans , Immediate-Early Proteins/genetics , Immunohistochemistry/methods , In Situ Hybridization/methods , Intercellular Signaling Peptides and Proteins/genetics , Male , Neoplasm Proteins/genetics , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
Swiss Surg ; 9(4): 193-7, 2003.
Article in English | MEDLINE | ID: mdl-12974179

ABSTRACT

BACKGROUND: There is accumulating evidence, both quantitative and qualitative, that pelvic irradiation affects anorectal function. However, the molecular mechanisms responsible for radiation-induced damage to the anal sphincter remain unclear. AIM: To determine the expression of transforming growth factor-beta 1 (TGF-beta 1) and its downstream effector connective tissue growth factor (CTGF) in the anal sphincter of a patient irradiated for prostate cancer. PATIENT: A 82 year-old patient developed a rectal adenocarcinoma and underwent an abdomino-perineal resection (APR), four years after receiving pelvic irradiation for prostate carcinoma. METHODS: Tissue sections of the anal sphincter were processed for histology. Immunostaining for TGF-beta 1 and CTGF were performed. RESULTS: CTGF and TGF-beta 1 immunoreactivity was detected in the irradiated anal sphincter, and was absent in controls. Immunoreactivity for both cytokines predominated in the internal sphincter. CTGF and TGF-beta 1 were preferentially detected in endothelial cells, myofibroblasts and fibroblasts; in addition, there was strong immunoreactivity for TGF-beta 1, but not for CTGF in smooth muscle cells of the anal canal. CONCLUSION: Four years after pelvic irradiation, radiation-induced damage appeared to affect predominantly the smooth muscle layer of the anal canal. The molecular mechanisms responsible for radiation-induced fibrosis to these tissues involve prolonged activation of TGF-beta 1 and its downstream effector CTGF.


Subject(s)
Adenocarcinoma/radiotherapy , Adenocarcinoma/surgery , Anal Canal/radiation effects , Immediate-Early Proteins/analysis , Intercellular Signaling Peptides and Proteins/analysis , Muscle, Smooth/radiation effects , Neoplasms, Radiation-Induced/surgery , Neoplasms, Second Primary/surgery , Prostatic Neoplasms/radiotherapy , Radiation Injuries/pathology , Rectal Neoplasms/surgery , Transforming Growth Factor beta/analysis , Aged , Aged, 80 and over , Anal Canal/pathology , Connective Tissue Growth Factor , Disease Progression , Fibrosis/pathology , Fibrosis/surgery , Humans , Immunoenzyme Techniques , Male , Muscle, Smooth/pathology , Rectum/pathology , Rectum/radiation effects , Rectum/surgery , Reoperation , Transforming Growth Factor beta1
5.
J Endocrinol ; 178(2): 169-75, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12904165

ABSTRACT

The CCN family comprises cysteine-rich 61 (CYR61/CCN1), connective tIssue growth factor (CTGF/CCN2), nephroblastoma overexpressed (NOV/CCN3), and Wnt-induced secreted proteins-1 (WISP-1/CCN4), -2 (WISP-2/CCN5) and -3 (WISP-3/CCN6). These proteins stimulate mitosis, adhesion, apoptosis, extracellular matrix production, growth arrest and migration of multiple cell types. Many of these activities probably occur through the ability of CCN proteins to bind and activate cell surface integrins. Accumulating evidence supports a role for these factors in endocrine pathways and endocrine-related processes. To illustrate the broad role played by the CCN family in basic and clinical endocrinology, this Article highlights the relationship between CCN proteins and hormone action, skeletal growth, placental angiogenesis, IGF-binding proteins and diabetes-induced fibrosis.


Subject(s)
Estrogens/metabolism , Immediate-Early Proteins/metabolism , Integrins/metabolism , Bone and Bones/metabolism , Breast Neoplasms/metabolism , CCN Intercellular Signaling Proteins , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Diabetes Mellitus, Type 1/metabolism , Female , Fibrosis , Growth Substances/metabolism , Humans , Insulin-Like Growth Factor Binding Proteins , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins , Neoplasm Proteins/metabolism , Neovascularization, Physiologic , Nephroblastoma Overexpressed Protein , Oncogene Proteins/metabolism , Osteogenesis/physiology , Placental Circulation , Pregnancy , Protein Binding , Proto-Oncogene Proteins , Repressor Proteins , Somatomedins/metabolism , Transcription Factors/metabolism , Uterine Neoplasms/metabolism
6.
Mol Pathol ; 56(2): 80-5, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12665625

ABSTRACT

For the second time, researchers from leading laboratories in the CCN field gathered in Saint-Malo, France, to participate in the Second International Workshop on the CCN family of genes. In addition to the regular research communications, meeting highlights included the inauguration of the first CCN newsletter (http://ccnnewsletter.free.fr) and the recognition of the International CCN Society (http://www.ccnsociety.jussieu.fr) as an important medium for the exchange of scientific knowledge and resources in the CCN field. Once more, the high quality of scientific communications and individual interactions set the stage for an extremely fruitful meeting.


Subject(s)
Immediate-Early Proteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Animals , Biomarkers, Tumor/metabolism , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Embryonic and Fetal Development/genetics , Gene Expression Regulation , Humans , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins/genetics , Mice , Neoplasm Proteins/metabolism , Neoplasms/genetics , Signal Transduction/physiology
7.
Mol Pathol ; 56(2): 127-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12665631

ABSTRACT

A proposal is put forth to unify the nomenclature of the CCN family of secreted, cysteine rich regulatory proteins. In the order of their description in the literature, CCN1 (CYR61), CCN2 (CTGF), CCN3 (NOV), CCN4 (WISP-1), CCN5 (WISP-2), and CCN6 (WISP-3) constitute a family of matricellular proteins that regulate cell adhesion, migration, proliferation, survival, and differentiation, at least in part through integrin mediated mechanisms. This proposal is endorsed by the International CCN Society and will serve to eliminate confusion from the multiple names that have been given to these molecules.


Subject(s)
Immediate-Early Proteins , Intercellular Signaling Peptides and Proteins , Terminology as Topic , Connective Tissue Growth Factor , Cysteine-Rich Protein 61 , Humans , Nephroblastoma Overexpressed Protein , Societies, Scientific
8.
Reproduction ; 125(2): 271-84, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12578541

ABSTRACT

Connective tissue growth factor (CTGF) stimulates cell proliferation, migration, adhesion and extracellular matrix production, and functions in processes such as development, differentiation, angiogenesis, implantation, wound healing and fibrosis. CTGF is a 38 kDa protein that comprises four discrete structural modules (modules 1-4) but is susceptible to limited proteolysis in utero yielding bioactive isoforms that comprise either modules 3 and 4 (16-20 kDa) or module 4 (10 kDa). Here we report the development of a stable cell line, termed DB1, that was generated by transfecting cDNA encoding full-length human CTGF into Chinese hamster ovary cells that were mutant for heparin sulphate and chondroitin sulphate. DB1 cells produced 38 kDa CTGF and low molecular mass CTGFs that had N-termini between modules 2 and 3 at Ala(181) (20 kDa), Leu(184) (18 kDa) or Ala(197) (16 kDa) or between modules 3 and 4 at Gly(253) (10 kDa). CTGF was exported from DB1 cells as early as 5 min after synthesis and all isoforms were readily purified from conditioned medium by sequential steps of heparin affinity, cation exchange, and reverse-phase chromatography. The 38 kDa CTGF was faithfully glycosylated and underwent limited proteolysis in the presence of thrombin, kallikrein or uterine fluids, the last of which was antagonized by anti-thrombin III. All CTGF isoforms promoted cell adhesion, mitosis and epithelial transdifferentiation in vitro as well as subcutaneous fibrosis in vivo. The establishment of this recombinant expression system allows for mass-scale production of all previously reported uterine CTGF isoforms, demonstrates that module 4 contains functional domains involved in a broad range of biological activities, and will facilitate studies of CTGF processing in vitro.


Subject(s)
Bioreactors , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/biosynthesis , Protein Isoforms/biosynthesis , Animals , CHO Cells , Chondroitin Sulfates , Connective Tissue Growth Factor , Cricetinae , Female , Glycosylation , Heparitin Sulfate , Humans , Immediate-Early Proteins/analysis , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/analysis , Intercellular Signaling Peptides and Proteins/metabolism , Protein Isoforms/analysis , Protein Isoforms/metabolism , Recombinant Proteins , Transfection , Uterus/metabolism
9.
J Endocrinol ; 176(2): R1-7, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12553878

ABSTRACT

Connective tissue growth factor (CTGF) is a 349-residue mosaic protein that contains four structural modules implicated in protein-protein interactions. To address the functionality of residues 247-349 (containing module 4 alone), this region of CTGF was produced as a maltose binding protein (MBP) fusion protein in E. coli. After removal of MBP, recombinant CTGF commenced at Glu(247), was of M(r) 10 000, was immunoreactive with anti-CTGF[247-260], bound strongly to heparin, and promoted dose-dependent adhesion of fibroblasts, myofibroblasts, endothelial cells, and epithelial cells. An 8 kDa presumptive C-terminally truncated form of CTGF commencing at Glu(247) also promoted cell adhesion. CTGF-mediated cell adhesion was abolished by heparin or EDTA. These data demonstrate the presence of heparin-binding and cell-adhesion motifs within the C-terminal 103 residues of CTGF and show that CTGF-mediated cell adhesion is heparin-and divalent cation-dependent. Thus, CTGF isoforms comprising essentially module 4 are intrinsically functional in the absence of the other constituent modules of CTGF.


Subject(s)
Connective Tissue Cells/cytology , Heparin/metabolism , Immediate-Early Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , 3T3 Cells/cytology , 3T3 Cells/drug effects , Animals , Anticoagulants/pharmacology , Cell Adhesion/drug effects , Connective Tissue Cells/drug effects , Connective Tissue Growth Factor , Dose-Response Relationship, Drug , Edetic Acid/pharmacology , Endothelium/cytology , Endothelium/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Heparin/pharmacology , Humans , Immediate-Early Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Liver/cytology , Liver/drug effects , Mice , Peptide Fragments/pharmacology , Protein Binding , Recombinant Proteins/pharmacology , Stimulation, Chemical
10.
Mol Pathol ; 55(3): 186-92, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032230

ABSTRACT

AIMS: To determine the localisation and distribution of connective tissue growth factor (CCN2; CTGF) and transforming growth factor beta type 1 (TGF-beta1) in uterine tissues from cycling and early pregnant pigs. METHODS: In situ hybridisation and immunohistochemistry were used to localise CCN2 (CTGF) or TGF-beta1 in uteri obtained from gilts on days 0, 5, 10, 12, 15, and 18 of the oestrous cycle or days 10, 12, 14, 16, 17, and 21 of gestation. RESULTS: In cycling animals, CCN2 (CTGF) mRNA and protein were abundant in luminal epithelial cells (LECs) and glandular epithelial cells (GECs), with lesser amounts in stromal fibroblasts and little or none in endothelial cells. A similar pattern of staining was seen up to day 10 of pregnancy, except that overall staining intensities for CCN2 (CTGF) mRNA or protein were higher and that stromal and endothelial cells were CCN2 (CTGF) positive. However, on days 12-17 there was a striking decrease in the amount of CCN2 (CTGF) in LECs at the utero-conceptus interface, which was associated with maternal stromal matrix reorganisation and the onset of subepithelial neovascularisation. This differential distribution of CCN2 (CTGF) was localised to those LECs that were in close proximity to or in apposition with trophoblast cells. This decrease in CCN2 (CTGF) staining was transient in nature and high amounts of CCN2 (CTGF) were again apparent in LECs on days 17-21, when endometrial neovascularisation and matrix remodelling were complete. The expression of uterine TGF-beta1 was comparable to that of CCN2 (CTGF) at most stages of the oestrous cycle or early pregnancy. Pre-elongation blastocysts recovered on day 10 were positive for both CCN2 (CTGF) and TGF-beta1 in the extra-embryonic trophectoderm, endoderm, and inner cell mass. On day 12, trophectoderm expressed low amounts of TGF-beta1 mRNA and non-detectable amounts of TGF-beta1 protein or CCN2 (CTGF) mRNA or protein. By days 17-21, the expression of both growth factors in the extra-embyronic/placental membranes increased and frequently exceeded that seen in LECs. CONCLUSIONS: The pattern of CCN2 (CTGF) production during the initial attachment phase supports a role for this factor in stromal remodelling and neovascularisation, although alternative functions at later stages such as epithelial-epithelial interactions are also possible. In most major cell types in the uterus or utero-placental unit, CCN2 (CTGF) expression was highly correlated with that of TGF-beta(1), indicating that CCN2 (CTGF) may mediate some of the functions of TGF-beta in the reproductive tract during the oestrous cycle and pregnancy. The data further highlight epithelium as an important source of CCN2 (CTGF) in the regulation of uterine function.


Subject(s)
Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Placenta/metabolism , Pregnancy/metabolism , Swine/metabolism , Transforming Growth Factor beta/biosynthesis , Uterus/metabolism , Animals , Connective Tissue Growth Factor , Estrous Cycle/physiology , Female , Gene Expression Regulation, Developmental , Growth Substances/genetics , Immediate-Early Proteins/genetics , In Situ Hybridization , RNA, Messenger/genetics , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1
11.
Mol Pathol ; 54(5): 338-46, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11577177

ABSTRACT

AIMS: To determine mechanisms regulating the production of connective tissue growth factor (CCN2; CTGF) and transforming growth factor beta1 (TGF-beta1) in the mouse uterus. METHODS: In situ hybridisation and immunohistochemistry were used to localise CCN2 (CTGF) and TGF-beta1 in uteri from sexually mature female mice that had either been (1) mated with sterile males to induce pseudopregnancy or (2) ovariectomised (OVX) and administered estradiol-17beta (E2) or progesterone (P4), either alone or in combination. Uteri collected on days 0.5, 1.5, 2.5, 3.5, 4.5, or 5.5 of pseudopregnancy or at one, three, six, 12, or 24 hours after steroid administration were fixed, sectioned, and incubated with specific riboprobes or antibodies to permit detection and localisation of mRNA or protein for CTGF and TGF-beta1. RESULTS: On days 0.5-2.5 of pseudopregnancy, CCN2 (CTGF) and TGF-beta1 were principally colocalised to uterine epithelial cells, with much smaller amounts in the stroma. On days 3.5-4.5, there was a reduction of CCN2 (CTGF) and TGF-beta1 in the epithelium but an increase in stromal and endothelial cells, corresponding to a period of extracellular matrix remodelling and neovascularisation within the endometrium. In OVX mice, epithelial cells were weakly positive for both CCN2 (CTGF) and TGF-beta1 in the absence of steroid hormones. Epithelial CTGF mRNA production were strongly but transiently stimulated in OVX mice cells by E2. These effects were antagonised by P4, which itself transiently stimulated epithelial CCN2 (CTGF) production, although less robustly than E2. CTGF and TGF-beta1 protein amounts were high in epithelial cells throughout steroid treatment and were increased in the stroma, where they were relatively long lived. Stromal CCN2 (CTGF) and TGF-beta1 were lower after co-administration of E2 and P4 than in response to each hormone individually. Although ccn2 (ctgf) is a TGF-beta1 inducible gene in other systems, and both growth factors were often co-localised in uterine tissues in these studies, several treatment regimens resulted in high amounts of TGF-beta1 protein in stromal cells without the concomitant production of ccn2 (ctgf) mRNA. CONCLUSIONS: Maternal factors are principal cues for CCN2 (CTGF) and TGF-beta1 production in the uterus because (1) their expression during pseudopregnancy is comparable to that seen in pregnancy and (2) they are regulated by ovarian steroids. TGF-beta dependent and independent mechanisms of ccn2 (ctgf) gene transcription exist in the uterus that are variably regulated by steroid hormones. Collectively, the data support a role for CCN2 (CTGF) in mediating the effects of steroid hormones and TGF-beta on endometrial function.


Subject(s)
Estradiol/physiology , Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Progesterone/physiology , Uterus/metabolism , Animals , Connective Tissue Growth Factor , Extracellular Matrix/physiology , Female , Immunohistochemistry , In Situ Hybridization , Mice , Neovascularization, Physiologic/physiology , Pseudopregnancy/physiopathology , RNA, Messenger/analysis , Transforming Growth Factor beta/biosynthesis
12.
Growth Factors ; 19(2): 127-43, 2001.
Article in English | MEDLINE | ID: mdl-11769972

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is a 22 kDa, O-glycosylated protein. HeLa cells infected with a recombinant vaccinia virus expressing human HB-EGF produced a secreted, bioactive protein, with Mr 22,000 that was decreased to 14,000 by treatment with O-glycanase. Site-directed mutagenesis of HB-EGF cDNA using oligonucleotide- and PCR-directed techniques was performed to change the potential glycosylation sites, Thr75 and Thr85, to alanine residues to prevent O-glycosylation. Purification and characterization of the mutant proteins demonstrated that: (i) both O-glycosylation sites of HB-EGF are utilized, (ii) HB-EGF secretion does not require O-glycosylation, (iii) removal of O-glycans does not affect proteolytic cleavage of the HB-EGF precursor, nor does it influence HB-EGF intracellular trafficking or subcellular localization, and (iv) HB-EGF produced by HeLa cells is heavily sialylated. Comparisons between glycosylation mutants and wild-type HB-EGF revealed no significant apparent differences in receptor binding activity.


Subject(s)
Epidermal Growth Factor/chemistry , Epidermal Growth Factor/genetics , Alanine/chemistry , Binding Sites , Blotting, Western , Chromatography, High Pressure Liquid , DNA, Complementary/metabolism , Electrophoresis, Polyacrylamide Gel , Epidermal Growth Factor/metabolism , Genetic Vectors , Glycosylation , HeLa Cells , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Mutation , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Threonine/chemistry , Vaccinia virus/metabolism
13.
Mol Hum Reprod ; 6(12): 1093-8, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11101692

ABSTRACT

Connective tissue growth factor (CTGF) is a recently described heparin-binding mitogen for fibroblasts and smooth muscle cells. The aim of this study was to investigate the production of CTGF by human uterine tissues using immunohistochemical and Northern blotting analyses. For immunohistochemistry, formalin-fixed human proliferative (n = 5), early secretory (n = 5; days 15-19), mid-secretory (n = 5; days 20-23), late secretory (n = 5; days 24-28) endometrial, and decidual (n = 5) tissues were stained using a highly specific affinity-purified polyclonal antibody raised against residues 81-94 of human CTGF. Myometrial (n = 5) and leiomyoma (n = 5) tissues were also used for CTGF immunochemistry. In proliferative endometrium, epithelial and vascular endothelial cells showed strong CTGF immunoreactivity, whereas stromal cells were negative or only weakly positive for the CTGF protein. Throughout the entire secretory stage, CTGF was detected in epithelial and endothelial cells of endometrium. Stromal cells showed strong immunoreactivity to CTGF only in oedematous areas for early and mid-secretory endometrium, and in decidualized regions of late secretory endometrium. During pregnancy, the decidual, epithelial and endothelial cells of the endometrium were all immunoreactive to CTGF. In myometrial and leiomyoma samples, CTGF immunoreactivity was found only in the endothelial cells. Northern blotting of mRNA from normal uterus (n = 2) or leiomyoma (n = 6) using a 320 bp human CTGF cDNA probe revealed a single 2.4 kb transcript. This study is the first to demonstrate CTGF gene expression and localization of its encoded protein in human uterine tissues. The cell- and cycle-specific localization of CTGF support a role for this molecule in regulating aspects of uterine cell growth, migration, and/or matrix production during the menstrual cycle and pregnancy.


Subject(s)
Growth Substances/analysis , Immediate-Early Proteins/analysis , Intercellular Signaling Peptides and Proteins , Mitogens/analysis , Uterus/chemistry , Adult , Blotting, Northern/methods , Connective Tissue Growth Factor , Decidua/chemistry , Decidua/pathology , Endometrium/chemistry , Endometrium/pathology , Female , Growth Substances/genetics , Humans , Immediate-Early Proteins/genetics , Immunoenzyme Techniques , Pregnancy , Uterus/pathology
14.
Mol Genet Metab ; 71(1-2): 276-92, 2000.
Article in English | MEDLINE | ID: mdl-11001822

ABSTRACT

Connective tissue growth factor (CTGF) is a member of the recently described CCN gene family which contains CTGF itself, cyr61, nov, elm1, Cop1, and WISP-3. CTGF is transcriptionally activated by several factors although its stimulation by transforming growth factor beta (TGF-beta) has attracted considerable attention. CTGF acts to promote fibroblast proliferation, migration, adhesion, and extracellular matrix formation, and its overproduction is proposed to play a major role in pathways that lead to fibrosis, especially those that are TGF-beta-dependent. This includes fibrosis of major organs, fibroproliferative diseases, and scarring. CTGF also appears to play a role in the extracellular matrix remodeling that occurs in normal physiological processes such as embryogenesis, implantation, and wound healing. However, recent advances have shown that CTGF is involved in diverse autocrine or paracrine actions in several other cell types such as vascular endothelial cells, epithelial cells, neuronal cells, vascular smooth muscle cells, and cells of supportive skeletal tissues. Moreover, in some circumstances CTGF has negative effects on cell growth in that it can be antimitotic and apoptotic. In light of these discoveries, CTGF has been implicated in a diverse variety of processes that include neovascularization, transdifferentiation, neuronal scarring, atherosclerosis, cartilage differentiation, and endochondral ossification. CTGF has thus emerged as a potential important effector molecule in both physiological and pathological processes and has provided a new target for therapeutic intervention in fibrotic diseases.


Subject(s)
Connective Tissue/physiology , Growth Substances/chemistry , Growth Substances/genetics , Immediate-Early Proteins/chemistry , Immediate-Early Proteins/genetics , Intercellular Signaling Peptides and Proteins , Amino Acid Sequence , Animals , Connective Tissue/growth & development , Connective Tissue Growth Factor , Endothelium, Vascular/physiology , Epithelium/physiology , Gene Expression , Growth Substances/physiology , Humans , Immediate-Early Proteins/physiology , Molecular Sequence Data , Muscle, Smooth/physiology , Nephroblastoma Overexpressed Protein , Nerve Tissue/physiology , Sequence Homology, Amino Acid , Terminology as Topic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/physiology
15.
Biol Reprod ; 63(2): 390-400, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10906042

ABSTRACT

We recently showed that insulin-like growth factor-binding protein-4 (IGFBP-4) proteolytic degradation in ovine preovulatory ovarian follicles is IGF-dependent and regulated by the heparin-binding domain (HBD) from IGFBP-3 and from connective tissue growth factor (CTGF), heparan/heparin-interacting protein (HIP), and vitronectin. The present study investigated regulation of IGFBP-4 proteolytic degradation in porcine, bovine, and equine ovarian preovulatory follicles. Follicular fluid from such preovulatory follicles contains proteolytic activity, degrading exogenous IGFBP-4. An excess of IGF-I enhanced IGFBP-4 degradation. In contrast, IGFBP-2 or -3 or monoclonal antibodies against IGF-I or -II dose-dependently inhibited IGFBP-4 degradation, and IGF-I or -II reversed this inhibition in a dose-dependent manner. Heparin-binding peptides derived from the C-terminal domain of IGFBP-3 or -5 inhibited IGFBP-4 degradation. Other heparin-binding peptides derived from CTGF, HIP, and vitronectin also inhibited IGFBP-4 degradation, except in porcine follicles. Finally, IGFBP-3 that was mutated in its HBD was less effective at inhibiting IGFBP-4 degradation. Thus, in bovine, porcine, and equine preovulatory follicles, IGFBP-4 proteolytic degradation both depends on IGFs and is inhibited by peptides containing HBD. Overall, these results suggest that during terminal development of follicles to the preovulatory stage in domestic animal species, the increase in IGF bioavailability might enhance IGFBP-4 degradation. In contrast, in atretic follicles, the decrease in IGF bioavailability, resulting partly from the increase in IGFBP-2 (sow, heifer, mare) and IGFBP-5 (heifer) expression would participate in the decrease of IGFBP-4 degradation. In bovine atretic follicles, IGFBP-5 would also strengthen the inhibition of IGFBP-4 degradation by direct interaction of its HBD with the protease. The involvement of other HBD-containing proteins in the modulation of intrafollicular proteases degrading IGFBP-4 remains to be investigated.


Subject(s)
Blood Coagulation Factors , Endopeptidases/metabolism , Heparin/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Intercellular Signaling Peptides and Proteins , Ovarian Follicle/enzymology , Ovulation , Somatomedins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Binding Sites , Carrier Proteins/pharmacology , Cattle , Connective Tissue Growth Factor , Female , Follicular Fluid/enzymology , Growth Substances/pharmacology , Horses , Immediate-Early Proteins/pharmacology , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor Binding Protein 5/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Peptide Fragments/pharmacology , RNA-Binding Proteins , Ribosomal Proteins , Swine , Vitronectin/pharmacology
16.
J Hepatol ; 32(5): 754-61, 2000 May.
Article in English | MEDLINE | ID: mdl-10845662

ABSTRACT

BACKGROUND/AIMS: Connective tissue growth factor is a recently described mitogenic protein implicated in a variety of fibrotic disorders. Connective tissue growth factor may be a downstream mediator of the pro-fibrotic and mitogenic actions of transforming growth factor-beta, promoting extracellular matrix deposition and fibrogenesis. As transforming growth factor-beta is considered important to the pathogenesis of hepatic fibrosis, we examined the possible contribution of connective tissue growth factor to this process. METHODS: Connective tissue growth factor expression was examined in normal and fibrotic human and rat livers using RT-PCR and ribonuclease protection assays, and in primary cultures of rat hepatic stellate cells by Northern and Western blotting. RESULTS: Ribonuclease protection assays demonstrated connective tissue growth factor mRNA was increased 3-5-fold in human fibrotic liver compared with normal. RT-PCR showed this mRNA was increased in carbon-tetrachloride-treated rat liver. Northern analysis showed connective tissue growth factor mRNA was increasingly expressed during progressive activation of cultured rat hepatic stellate cells. Western analysis confirmed that freshly isolated hepatic stellate cells secreted relatively little connective tissue growth factor compared with hepatic stellate cells activated in culture. Hepatic stellate cells stimulated with transforming growth factor-beta showed increased expression of connective tissue growth factor mRNA and protein. CONCLUSIONS: Connective tissue growth factor mRNA is consistently upregulated in human liver cirrhosis of various aetiologies, supporting a role for this growth factor in hepatic fibrogenesis. Our studies suggest that hepatic stellate cells may be an important source of hepatic connective tissue growth factor in vivo, particularly following stimulation with transforming growth factor-beta.


Subject(s)
Connective Tissue Cells/metabolism , Growth Substances/biosynthesis , Immediate-Early Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins , Liver Cirrhosis, Experimental/metabolism , Liver Cirrhosis/metabolism , Animals , Cells, Cultured , Connective Tissue Cells/pathology , Connective Tissue Growth Factor , Humans , Liver/metabolism , Liver/pathology , RNA, Messenger/analysis , Rats
17.
Growth Factors ; 17(2): 115-24, 1999.
Article in English | MEDLINE | ID: mdl-10595311

ABSTRACT

Connective tissue growth factor (CTGF) is a 38 kDa modular protein that is involved in processes such as cell proliferation, survival, migration, adhesion and extracellular matrix production. Target cells for CTGF include fibroblasts, smooth muscle cells and endothelial cells. Using a specific peptide antibody, CTGF was localized in tissue sections obtained from mouse embryos between days 7.5 and 14.5 of gestation. On day 7.5, CTGF was present at high levels in the embryonic ectoderm, mesoderm, and chorion, with weaker levels in the squamous endoderm. No CTGF was detected in Reichert's membrane, parietal endoderm, and visceral endoderm. Decidual cells of the maternal uterus stained strongly for CTGF. There was no specific staining for CTGF on day 11.5 but by day 13.5, the protein was detectable in a limited number of structures, most notably the liver, thymus, lung, and intestine. By day 14.5, staining for CTGF had become extensive and was present at high or moderate levels in the thymus, aorta, trachea, liver, lung, adrenal gland, kidney, iliac sinus, olfactory region, tongue, pharynx, esophagus, thyroglossal duct, choroid plexi, Rathke's pouch, stomach, pancreas, and midgut. Relatively weak staining was present in the heart and dorsal root ganglia. There was no staining in the vitelline duct or in the outer cortical or medullary regions of the brain. Among specific cell types, CTGF was localized at high levels in secretory and absorptive epithelial cells and liver parenchyma cells, at moderate levels in vascular endothelial cells and myoblasts and at low levels in mesenchymal and connective tissue cells. These data show that various tissues and organ systems produce CTGF in a specific temporo-spatial pattern during embryogenesis and support a role for CTGF in cellular differentiation and development during prenatal life.


Subject(s)
Embryo, Mammalian/chemistry , Growth Substances/analysis , Immediate-Early Proteins , Intercellular Signaling Peptides and Proteins , Animals , Connective Tissue Growth Factor , Embryonic and Fetal Development , Female , Gestational Age , Immunohistochemistry , Mice , Molecular Weight
18.
Growth Factors ; 17(1): 49-61, 1999.
Article in English | MEDLINE | ID: mdl-10495962

ABSTRACT

Heparin-binding epidermal growth factor-like growth factor (HB-EGF) is initially synthesized as a membrane bound protein that is subsequently processed to yield an approximately 74 amino acid secreted product. To investigate the biological activities of HB-EGF and its role(s) in tumor formation, the full-length HB-EGF cDNA was cloned under the regulation of the mouse metallothionein promoter and stably expressed in HB-EGF deficient mouse L cells. HB-EGF immunoreactive proteins of 21 and 24 kDa were observed from transfected MLC lysates, and these lysates exhibited the ability to bind to the EGF receptor, stimulate 3H-thymidine uptake in BALB/c-3T3 cells, and induce anchorage independent growth (AIG) of normal rat kidney (NRK) cells. Furthermore, NRK cells treated with either E. coli-derived or vaccinia virus-derived HB-EGF, as well as NRK cells directly transfected with the HB-EGF construct, demonstrated AIG. We conclude that HB-EGF is a potent growth factor capable of stimulating altered cell growth and anchorage independence.


Subject(s)
Epidermal Growth Factor/physiology , Heparin , 3T3 Cells , Animals , Blotting, Northern , Blotting, Western , Carcinogenicity Tests , Cell Division , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fluorescent Antibody Technique, Indirect , Heparin/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , L Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Transfection
19.
Endocrinology ; 140(9): 4175-84, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10465290

ABSTRACT

The regulation of insulin-like growth factor binding protein (IGFBP)-4 proteolytic degradation by insulin-like growth factors (IGFs) has been largely studied in vitro, but not in vivo. The aim of this study was to investigate the involvement of IGFs, IGFBP-2, IGFBP-3, and IGFBP-3 proteolytic fragments in the regulation of IGFBP-4 proteolytic activity in ovine ovarian follicles. Follicular fluid from preovulatory follicles contains proteolytic activity degrading exogenous IGFBP-4. The addition of an excess of IGF-I enhanced IGFBP-4 proteolytic degradation, whereas addition of IGFBP-2 or -3 or monoclonal antibodies against IGF-I and -II dose dependently inhibited IGFBP-4 proteolytic degradation. IGF-I and IGF-II, but not LongR3-IGF-I, reversed this inhibition in a dose-dependent manner. C-terminal, but not N-terminal, proteolytic fragments derived from IGFBP-3 (aa 161-264), as well as heparin-binding domain-containing peptides derived from the C-terminal domain of IGFBP-3 and -5 also induced the inhibition of IGFBP-4 proteolytic degradation. Other heparin-binding domain-containing peptides derived from the connective tissue growth factor (CTGF) and from proteins not related to IGFBP, heparan/heparin interacting protein (HIP) and vitronectin, but not from p36 subunit of annexin II tetramer, inhibited IGFBP-4 degradation. Furthermore, IGFBP-3, mutated on its heparin-binding domain, was not able to inhibit IGFBP-4 proteolytic degradation. So, in ovine preovulatory follicles, IGFBP-4 proteolytic degradation both 1) depends on IGFs, and 2) is inhibited by IGFBP-3 via its C-terminal heparin-binding domain as well as by heparin-binding domain containing peptides. These data suggest that in early atretic follicles, the increase in IGFBP-2 participates in the decrease in IGFBP-4 degradation. In late atretic follicles, the increase in the levels of C-terminal IGFBP-3 proteolytic fragments, generated by IGFBP-3 degradation, as well as the increase in IGFBP-5 expression would strengthen the inhibition of IGFBP-4 degradation. This inhibition might be partly mediated by direct interaction of IGFBP-4 proteinase(s) and heparin-binding domain within the C-terminal region from IGFBP-3 and -5.


Subject(s)
Follicular Phase/physiology , Insulin-Like Growth Factor Binding Protein 4/metabolism , Ovarian Follicle/metabolism , Peptide Hydrolases/metabolism , Animals , Female , Heparin/metabolism , Insulin-Like Growth Factor Binding Protein 3/genetics , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 3/pharmacology , Insulin-Like Growth Factor I/pharmacology , Mutation/physiology , Peptide Fragments/pharmacology , Peptides/chemical synthesis , Peptides/pharmacology , Sheep , Somatomedins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...