Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Aging Cell ; 20(3): e13313, 2021 03.
Article En | MEDLINE | ID: mdl-33561324

Pregnancy-associated plasma protein-A (PAPP-A) is a secreted metalloprotease that increases insulin-like growth factor (IGF) availability by cleaving IGF-binding proteins. Reduced IGF signaling extends longevity in multiple species, and consistent with this, PAPP-A deletion extends lifespan and healthspan; however, the mechanism remains unclear. To clarify PAPP-A's role, we developed a PAPP-A neutralizing antibody and treated adult mice with it. Transcriptomic profiling across tissues showed that anti-PAPP-A reduced IGF signaling and extracellular matrix (ECM) gene expression system wide. The greatest reduction in IGF signaling occurred in the bone marrow, where we found reduced bone, marrow adiposity, and myelopoiesis. These diverse effects led us to search for unifying mechanisms. We identified mesenchymal stromal cells (MSCs) as the source of PAPP-A in bone marrow and primary responders to PAPP-A inhibition. Mice treated with anti-PAPP-A had reduced IGF signaling in MSCs and dramatically decreased MSC number. As MSCs are (1) a major source of ECM and the progenitors of ECM-producing fibroblasts, (2) the originating source of adult bone, (3) regulators of marrow adiposity, and (4) an essential component of the hematopoietic niche, our data suggest that PAPP-A modulates bone marrow homeostasis by potentiating the number and activity of MSCs. We found that MSC-like cells are the major source of PAPP-A in other tissues also, suggesting that reduced MSC-like cell activity drives the system-wide reduction in ECM gene expression due to PAPP-A inhibition. Dysregulated ECM production is associated with aging and drives age-related diseases, and thus, this may be a mechanism by which PAPP-A deficiency enhances longevity.


Homeostasis , Longevity , Mesenchymal Stem Cells/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Animals , Antibodies, Neutralizing/metabolism , Bone Marrow/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Models, Biological , Myelopoiesis , Osteoblasts/metabolism , Osteogenesis , Pregnancy-Associated Plasma Protein-A/metabolism , Signal Transduction , Somatomedins/metabolism
2.
Biochem Biophys Res Commun ; 543: 29-37, 2021 03 05.
Article En | MEDLINE | ID: mdl-33508770

Pregnancy-associated plasma protein-A (PAPP-A), a type of metalloproteinase in the insulin-like growth factor (IGF) system, has been implicated in atherosclerosis progression, but its function and mechanism in atherosclerosis is not fully understood. The study was performed to further explore the effects of PAPP-A on inflammation, macrophage polarization and atherosclerosis. In mouse macrophages stimulated by oxidized low-density lipoprotein (ox-LDL), PAPP-A expression was significantly increased. Its knockdown markedly mitigated inflammatory response and polarized macrophages to an M2-like phenotype in RAW264.7 cells upon ox-LDL treatment. Additionally, ox-LDL-induced activation of nuclear factor-κB (NF-κB) signaling pathway was dramatically restricted by PAPP-A knockdown in macrophages. However, JAK2/STAT3 activation was significantly up-regulated in RAW264.7 cells with PAPP-A inhibition after ox-LDL treatment. Importantly, we found that PAPP-A knockdown-induced polarization of M2-like phenotype in macrophages was mainly dependent on STAT3 activation. Clinical studies showed that serum PAPP-A levels were higher in patients with coronary artery disease (CAD) than that of healthy individuals. Apolipoprotein E-knockout (ApoE-/-) mice with high fat diet (HFD)-induced atherosclerosis exhibited higher expression of PAPP-A in aortas, which was mainly colocalized with F4/80. Subsequently, we found that PAPP-A deficiency greatly alleviated plaque formation, lesion burden and collagen accumulation in HFD-fed ApoE-/- mice. Consistent with in vitro macrophage phenotype, PAPP-A-/- reduced F4/80 expression, NF-κB activation and inflammatory response, while improved janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signaling and polarized macrophages to an M2-like phenotype in aortas of ApoE-/- mice after HFD feeding. In conclusion, these findings identified PAPP-A as a positive regulator of atherosclerosis by regulating macrophage polarization via STAT3 signal, and thus could be considered as a potential therapeutic target for atherosclerosis treatment.


Apolipoproteins E/genetics , Atherosclerosis/prevention & control , Diet, High-Fat , Macrophages/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Humans , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction
3.
Endocrinology ; 161(10)2020 10 01.
Article En | MEDLINE | ID: mdl-32888014

Pathogenicity of visceral adipose tissue (VAT) has been linked to the metabolic stress of enlarging mature adipocytes and a limited ability to recruit new adipocytes. One of the major distinguishing features of VAT preadipocytes is the high expression of the zinc metalloprotease, pregnancy-associated plasma protein-A (PAPP-A), when compared to subcutaneous adipose tissue (SAT). In this study we used 2 different approaches to investigate the effect of PAPP-A inhibition on different fat depots in mice on a high-fat diet (HFD) for 15 weeks. Conditional knockdown of PAPP-A gene expression in female adult mice resulted in significant decreases of 30% to 40% in adipocyte size in VAT (mesenteric and pericardial depots) compared to control mice. There was no effect on SAT (inguinal) or intra-abdominal perigonadal fat. Liver lipid was also significantly decreased without any effect on heart and skeletal muscle lipid. We found similar effects when using a pharmacological approach. Weekly injections of a specific immunoneutralizing monoclonal antibody (mAb-PA 1/41) or isotype control were given to male and female wild-type mice on HFD for 15 weeks. Adipocyte size was significantly decreased (30%-50%) only in VAT with mAb-PA 1/41 treatment. In this model, cell number was significantly increased in mesenteric fat in mice treated with mAb-PA 1/41, suggesting hyperplasia along with reduced hypertrophy in this VAT depot. Gene expression data indicated a significant decrease in F4/80 (macrophage marker) and interleukin-6 (proinflammatory cytokine) and a significant increase in adiponectin (anti-inflammatory adipokine with beneficial metabolic effects) in mesenteric fat compared to inguinal fat in mice treated with mAb-PA 1/41. Furthermore, there was significantly decreased liver lipid content with mAb-PA 1/41 treatment. Thus, using 2 different models systems we provide proof of principle that PAPP-A inhibition is a potential therapeutic target to prevent visceral obesity and its metabolic sequelae, such as fatty liver.


Anti-Obesity Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Obesity, Abdominal , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Diet, High-Fat/adverse effects , Female , Gene Knockout Techniques , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity, Abdominal/genetics , Obesity, Abdominal/prevention & control , Pregnancy-Associated Plasma Protein-A/immunology , Subcutaneous Fat/metabolism
4.
Sci Rep ; 10(1): 13779, 2020 08 13.
Article En | MEDLINE | ID: mdl-32792532

Elevated levels of pregnancy-associated plasma protein-A (PAPP-A) have been implicated in the pathogenesis of various malignancies, including breast cancers. Breast cancer is one of the most frequent carcinomas and is the second most common cancer type detected in women of child-bearing age. Throughout pregnancy PAPP-A is produced and secreted by the placental syncytiotrophoblast cells; co-incidentally pregnancy-associated breast cancers often have an aggressive clinical course. The components of the PAPP-A/IGF axis was assessed in a panel of breast cancer cell lines. Using neutralising antibodies the impact of PAPP-A/IGF axis on cell motility was evaluated. PAPP-A was expressed in four of the twelve breast cancer cell lines tested. Blocking PAPP-A and IGFBP4 with neutralising antibodies significantly decreased motiliy of MDA-MB-231 cells. Upregulation of PAPP-A expression in breast tumours resulted in a trend towards worse overall survival. Notably, PAPP-A expression also positively correlated with epithelial-to-mesenchymal transition markers. In conclusion, these results indicate that PAPP-A plays an important role in breast cancer progression and it may be a promising therapeutic target in breast cancer.


Breast Neoplasms/pathology , Disease Progression , Insulin-Like Growth Factor Binding Protein 4/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition/physiology , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/antagonists & inhibitors , Insulin-Like Growth Factor I/metabolism , Neoplasm Invasiveness , Pregnancy , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Prognosis , Trophoblasts/metabolism
5.
Cell Mol Gastroenterol Hepatol ; 10(1): 113-132, 2020.
Article En | MEDLINE | ID: mdl-32004755

BACKGROUND & AIMS: In patients with autoimmune atrophic gastritis and achlorhydria, hypergastrinemia is associated with the development of type 1 gastric neuroendocrine tumors (gNETs). Twelve months of treatment with netazepide (YF476), an antagonist of the cholecystokinin B receptor (CCKBR or CCK2R), eradicated some type 1 gNETs in patients. We investigated the mechanisms by which netazepide induced gNET regression using gene expression profiling. METHODS: We obtained serum samples and gastric corpus biopsy specimens from 8 patients with hypergastrinemia and type 1 gNETs enrolled in a phase 2 trial of netazepide. Control samples were obtained from 10 patients without gastric cancer. We used amplified and biotinylated sense-strand DNA targets from total RNA and Affymetrix (Thermofisher Scientific, UK) Human Gene 2.0 ST microarrays to identify differentially expressed genes in stomach tissues from patients with type 1 gNETs before, during, and after netazepide treatment. Findings were validated in a human AGSGR gastric adenocarcinoma cell line that stably expresses human CCK2R, primary mouse gastroids, transgenic hypergastrinemic INS-GAS mice, and patient samples. RESULTS: Levels of pappalysin 2 (PAPPA2) messenger RNA were reduced significantly in gNET tissues from patients receiving netazepide therapy compared with tissues collected before therapy. PAPPA2 is a metalloproteinase that increases the bioavailability of insulin-like growth factor (IGF) by cleaving IGF binding proteins (IGFBPs). PAPPA2 expression was increased in the gastric corpus of patients with type 1 gNETs, and immunohistochemistry showed localization in the same vicinity as CCK2R-expressing enterochromaffin-like cells. Up-regulation of PAPPA2 also was found in the stomachs of INS-GAS mice. Gastrin increased PAPPA2 expression with time and in a dose-dependent manner in gastric AGSGR cells and mouse gastroids by activating CCK2R. Knockdown of PAPPA2 in AGSGR cells with small interfering RNAs significantly decreased their migratory response and tissue remodeling in response to gastrin. Gastrin altered the expression and cleavage of IGFBP3 and IGFBP5. CONCLUSIONS: In an analysis of human gNETS and mice, we found that gastrin up-regulates the expression of gastric PAPPA2. Increased PAPPA2 alters IGF bioavailability, cell migration, and tissue remodeling, which are involved in type 1 gNET development. These effects are inhibited by netazepide.


Benzodiazepinones/pharmacology , Neuroendocrine Tumors/drug therapy , Phenylurea Compounds/pharmacology , Pregnancy-Associated Plasma Protein-A/metabolism , Stomach Neoplasms/drug therapy , Animals , Benzodiazepines/pharmacology , Benzodiazepinones/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Gastric Mucosa/cytology , Gastric Mucosa/pathology , Gastrins/antagonists & inhibitors , Gastrins/blood , Gastrins/metabolism , Gene Knockdown Techniques , Humans , Mice , Mice, Transgenic , Neuroendocrine Tumors/blood , Neuroendocrine Tumors/pathology , Organoids , Phenylurea Compounds/therapeutic use , Pregnancy-Associated Plasma Protein-A/analysis , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Primary Cell Culture , Receptor, Cholecystokinin B/antagonists & inhibitors , Receptor, Cholecystokinin B/metabolism , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Treatment Outcome
6.
PLoS One ; 14(11): e0224564, 2019.
Article En | MEDLINE | ID: mdl-31751381

OBJECTIVES: Inhibition of pregnancy-associated plasma protein-A (PAPP-A), an upstream activator of the insulin-like growth factor (IGF) pathway, is known to augment sensitivity to platinum-based chemotherapy. This study further tests the efficacy of PAPP-A inhibition with a monoclonal antibody inhibitor (mAb-PA) in ovarian cancer (OC) platinum-resistant patient-derived xenograft (PDX) models. METHODS: PAPP-A expression was quantitated in platinum-resistant PDX models by ELISA. A subset with High (n = 5) and Low (n = 2) expression were revived in female SCID/beige mice for studies with either saline, carboplatin/paclitaxel (CP) + mAb-PA, or CP + IgG2a. The primary endpoint was tumor area by ultrasound on day 28 relative to baseline. Conversion to platinum-sensitive was defined by average tumor regression below baseline. Statistical analyses included linear mixed effects modeling and Kaplan Meier curves. Response to therapy was correlated with changes in the ratio of phosphorylated/total AKT and ERK 1/2 using Wes analysis. RESULTS: The addition of mAb-PA to CP induced tumor regression below baseline in one High PAPP-A PDX model; another three models exhibited notable growth inhibition relative to CP + IgG2a. None of the Low PAPP-A PDX models regressed below baseline. The PDX model with the greatest magnitude of tumor regression from baseline after combination therapy was maintained on single agent mAb-PA or IgG2a, but no benefit was observed. Decreased phosphorylation of ERK1/2 correlated with conversion to platinum-sensitive. CONCLUSIONS: The addition of mAb-PA to CP overcame platinum-resistance in one of five High PAPP-A PDX models; three other models demonstrated improved platinum-response. This supports further clinical development of this novel therapeutic.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/pharmacology , Carboplatin/therapeutic use , Cytoreduction Surgical Procedures , Female , Humans , Mice , Ovarian Neoplasms/pathology , Ovarian Neoplasms/surgery , Ovary/pathology , Ovary/surgery , Paclitaxel/therapeutic use , Xenograft Model Antitumor Assays
7.
Curr Vasc Pharmacol ; 15(3): 197-206, 2017.
Article En | MEDLINE | ID: mdl-28034366

Pregnancy-Associated Plasma Protein-A (PAPP-A) is a zinc-binding metalloproteinase protein produced by placental syncytio-trophoblasts and secreted into the maternal circulation where its concentration progressively increases until term. In recent years, PAPP-A has been studied for its potential involvement in cardiovascular (CV) disease. However, all those studies did not provide a clear view to identify the pathophysiological links between PAPP-A plasma levels and the occurrence of CV events. In this review, starting from a complete description of PAPP-A structure and biology, we present an updated overview of experimental as well as clinical evidence on the role of this metalloproteinase in CV disease. Finally, we discuss possible therapeutic approaches to antagonize its potential detrimental CV effects.


Cardiovascular Diseases/enzymology , Cardiovascular System/enzymology , Pregnancy-Associated Plasma Protein-A/metabolism , Animals , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/drug effects , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Female , Humans , Pregnancy , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/genetics , Protease Inhibitors/therapeutic use , Protein Conformation , Signal Transduction , Structure-Activity Relationship
8.
Hum Reprod ; 31(4): 866-74, 2016 Apr.
Article En | MEDLINE | ID: mdl-26874357

STUDY QUESTION: Is the proteolytic activity of pregnancy-associated plasma protein-A (PAPP-A) regulated by the stanniocalcins (STC1 and STC2) during human follicle maturation? SUMMARY ANSWER: The STCs and PAPP-A show similar expression by immunohistochemistry in developing follicles, and regulation of PAPP-A proteolytic activity is suggested by the identification of inhibited protein complexes between PAPP-A and STC1 or STC2 in human follicular fluid (FF). WHAT IS KNOWN ALREADY: The insulin-like growth factor (IGF)-regulating proteinase PAPP-A is secreted by the granulosa cells of estrogen-dominant follicles and is involved in follicle growth. STC1 and STC2 have recently been identified as novel PAPP-A inhibitors, and their expression in non-human mammalian ovaries has previously been observed. STUDY DESIGN, SIZE, DURATION: The proteolytic activity of PAPP-A in human follicular fluid was assessed, and the interaction between PAPP-A and the STCs in human ovarian tissues and follicular fluid was analyzed using immunoassays. From 21 women, matched pairs of follicular fluid were obtained from one follicle just prior to final maturation of follicles with human chorionic gonadotrophin (hCG), and from another follicle in connection with oocyte aspiration after hCG treatment. Ovarian tissues were obtained from women having one ovary removed for fertility preservation by cryopreservation prior to gonadotoxic treatment. PARTICIPANTS/MATERIALS, SETTING, METHODS: The concentration and activity of PAPP-A were determined in all samples of follicular fluid. Furthermore, to investigate PAPP-A regulation during follicle development, immunohistochemical staining of PAPP-A, STC1, and STC2 was performed on pre-antral and antral human follicles. To attempt the demonstration of native complexes between PAPP-A and the STCs, immunoprecipitation from a pool of human follicular fluid was performed. MAIN RESULTS AND THE ROLE OF CHANCE: The concentration of PAPP-A antigen in follicular fluid increased upon stimulation of ovulation with hCG (P < 0.02), but at the same time, PAPP-A activity was decreased. PAPP-A, STC1, and STC2 were localized together in primordial, late primary, and antral follicles, indicating that complex formation is possible in ovarian tissue. Covalent PAPP-A:STC2 and non-covalent PAPP-A:STC1 complexes were immunoprecipitated from follicular fluid, documenting for the first time native inhibited complexes between PAPP-A and the STCs. LIMITATIONS, REASONS FOR CAUTION: We have demonstrated the presence of native complexes between PAPP-A and the STCs in the human ovary, indicating STC-mediated PAPP-A proteolytic inhibition. Further investigation is required to extend this principle to other tissues. WIDER IMPLICATIONS OF THE FINDINGS: Our data suggest that the STCs contribute to PAPP-A regulation during folliculogenesis and support a general model in which STC1 and STC2 are regulators of mammalian IGF activity through inhibition of PAPP-A. We suggest that future functional studies take both PAPP-A and the STCs into consideration. STUDY FUNDING/COMPETING INTERESTS: This work was supported by grants from the Novo Nordisk Foundation, and the Danish Council for Independent Research. No competing interests declared.


Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Oogenesis , Ovarian Follicle/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Protease Inhibitors/metabolism , Adult , Chorionic Gonadotropin/genetics , Chorionic Gonadotropin/pharmacology , Female , Fertility Preservation , Follicular Fluid/drug effects , Follicular Fluid/enzymology , Follicular Fluid/metabolism , Glycoproteins/chemistry , Humans , Immunohistochemistry , Immunoprecipitation , Intercellular Signaling Peptides and Proteins/chemistry , Oogenesis/drug effects , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Ovulation Induction , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Protease Inhibitors/chemistry , Protein Interaction Domains and Motifs , Proteolysis/drug effects , Recombinant Proteins/pharmacology
9.
J Cardiovasc Transl Res ; 9(1): 77-9, 2016 Feb.
Article En | MEDLINE | ID: mdl-26733326

The metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), has been implicated in the development of cardiovascular disease in humans and mouse models. In the latter, genetic deletion or overexpression of PAPP-A confirmed a major role for PAPP-A in atherosclerosis. In this study, we tested the hypothesis that targeting PAPP-A proteolytic activity by an inhibitory monoclonal antibody (mAb-PA) reduces atherosclerotic plaque progression. Apolipoprotein E knock-out mice on high-fat diet were treated with mAb-PA or isotype control. Control mice had a 10-fold increase in aortic plaque after 10 weeks. Aortic plaque burden was reduced by ∼ 70% in mice treated with mAb-PA (P = 0.0002). Treatment was efficacious even in the face of elevated cholesterol and triglycerides. This study demonstrates proof-of-principle and provides feasibility for a novel therapeutic strategy to inhibit atherosclerotic plaque burden by selective targeting of PAPP-A.


Antibodies, Monoclonal/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Plaque, Atherosclerotic , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Protease Inhibitors/pharmacology , Animals , Aorta/enzymology , Aorta/immunology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/immunology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/immunology , Cholesterol/blood , Diet, High-Fat , Disease Models, Animal , Feasibility Studies , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Pregnancy-Associated Plasma Protein-A/immunology , Pregnancy-Associated Plasma Protein-A/metabolism , Time Factors , Triglycerides/blood
10.
J Biol Chem ; 290(36): 21915-24, 2015 Sep 04.
Article En | MEDLINE | ID: mdl-26195635

Stanniocalcin-1 (STC1) is a disulfide-bound homodimeric glycoprotein, first identified as a hypocalcemic hormone important for maintaining calcium homeostasis in teleost fish. STC1 was later found to be widely expressed in mammals, although it is not believed to function in systemic calcium regulation in these species. Several physiological functions of STC1 have been reported, although many molecular details are still lacking. We here demonstrate that STC1 is an inhibitor of the metzincin metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), which modulates insulin-like growth factor (IGF) signaling through proteolytic cleavage of IGF-binding proteins (IGFBPs). STC1 potently (Ki = 68 pm) inhibits PAPP-A cleavage of IGFBP-4, and we show in a cell-based assay that STC1 effectively antagonizes PAPP-A-mediated type 1 IGF receptor (IGF1R) phosphorylation. It has recently been found that the homologous STC2 inhibits PAPP-A proteolytic activity, and that this depends on the formation of a covalent complex between the inhibitor and the proteinase, mediated by Cys-120 of STC2. We find that STC1 is unable to bind covalently to PAPP-A, in agreement with the absence of a corresponding cysteine residue. It rather binds to PAPP-A with high affinity (KD = 75 pm). We further demonstrate that both STC1 and STC2 show inhibitory activity toward PAPP-A2, but not selected serine proteinases and metalloproteinases. We therefore conclude that the STCs are proteinase inhibitors, probably restricted in specificity to the pappalysin family of metzincin metalloproteinases. Our data are the first to identify STC1 as a proteinase inhibitor, suggesting a previously unrecognized function of STC1 in the IGF system.


Glycoproteins/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Blotting, Western , Glycoproteins/genetics , Glycoproteins/pharmacology , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 4/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Phosphorylation/drug effects , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Proteolysis/drug effects , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
11.
Mol Cancer Ther ; 14(4): 973-81, 2015 Apr.
Article En | MEDLINE | ID: mdl-25695953

The majority of ovarian cancer patients acquire resistance to standard platinum chemotherapy and novel therapies to reduce tumor burden and ascites accumulation are needed. Pregnancy-associated plasma protein-A (PAPP-A) plays a key role in promoting insulin-like growth factor (IGF) pathway activity, which directly correlates to ovarian cancer cell transformation, growth, and invasiveness. Herein, we evaluate PAPP-A expression in tumors and ascites of women with ovarian cancer, and determine the antitumor efficacy of a neutralizing monoclonal PAPP-A antibody (mAb-PA) in ovarian cancer using primary patient ovarian tumorgrafts ("Ovatars"). PAPP-A mRNA expression in patient ovarian tumors correlated with poor outcome and was validated as a prognostic surrogate in Ovatar tumors. Following confirmation of mAb-PA bioavailability and target efficacy in vivo, the antitumor efficacy of mAb-PA in multiple Ovatar tumor models was examined and the response was found to depend on PAPP-A expression. Strikingly, the addition of mAb-PA to standard platinum chemotherapy effectively sensitized platinum-resistant Ovatar tumors. PAPP-A protein in ascites was also assessed in a large cohort of patients and very high levels were evident across the entire sample set. Therefore, we evaluated targeted PAPP-A inhibition as a novel approach to managing ovarian ascites, and found that mAb-PA inhibited the development, attenuated the progression, and induced the regression of Ovatar ascites. Together, these data indicate PAPP-A as a potential palliative and adjunct therapeutic target for women with ovarian cancer.


Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , Antineoplastic Agents/administration & dosage , Ascites/pathology , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Gene Expression , Humans , Immunohistochemistry , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/metabolism , Prognosis , Xenograft Model Antitumor Assays
12.
Oncotarget ; 5(4): 1014-25, 2014 Feb 28.
Article En | MEDLINE | ID: mdl-24572990

The insulin-like growth factor (IGF) signaling pathway is involved in certain human cancers, and the feasibility of directly targeting the IGF receptor has been actively investigated. However, recent evidence from clinical trials suggests that this approach can be problematic. We have developed an alternative strategy to indirectly inhibit the IGF signaling by targeting the metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A). PAPP-A associated with the cell surface cleaves IGF binding protein-4 (IGFBP-4), when IGF is bound to IGFBP-4, and thereby increases IGF bioavailability for receptor activation in an autocrine/paracrine manner. We hypothesized that inhibition of PAPP-A would suppress excessive local IGF signaling in tissues where this is caused by increased PAPP-A proteolytic activity. To test this hypothesis, we developed an inhibitory monoclonal antibody, mAb 1/41, which targets a unique substrate-binding exosite of PAPP-A. This inhibitor selectively and specifically inhibits proteolytic cleavage of IGFBP-4 with an inhibitory constant (Ki) of 135 pM. In addition, it inhibited intracellular signaling of the IGF receptor (AKT phosphorylation) in monolayers of A549 cells, an IGF-responsive lung cancer-derived cell line found to express high levels of PAPP-A. We further showed that mAb 1/41 is effective towards PAPP-A bound to cell surfaces, and that it is capable of inhibiting PAPP-A activity in vivo. Using a murine xenograft model of A549 cells, we demonstrated that mAb 1/41 administered intraperitoneally significantly inhibited tumor growth. Analysis of xenograft tumor tissue recovered from treated mice showed penetration of mAb 1/41, reduced IGFBP-4 proteolysis, and reduced AKT phosphorylation. Our study provides proof of concept that IGF signaling can be selectively reduced by targeting a regulatory proteinase that functions extracellularly, upstream of the IGF receptor. PAPP-A targeting thus represents an alternative therapeutic strategy for inhibiting IGF receptor signaling.


Antibodies, Monoclonal/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Receptors, Somatomedin/metabolism , Animals , Antibodies, Monoclonal/immunology , Disease Models, Animal , Female , HEK293 Cells , Heterografts , Humans , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/enzymology , Male , Mice , Mice, Knockout , Molecular Targeted Therapy , Pregnancy , Pregnancy-Associated Plasma Protein-A/immunology , Pregnancy-Associated Plasma Protein-A/pharmacology , Signal Transduction , Transfection , Xenograft Model Antitumor Assays
13.
Biol Chem ; 388(5): 507-12, 2007 May.
Article En | MEDLINE | ID: mdl-17516846

Pregnancy-associated plasma protein-A (PAPP-A) is a metalloprotease that cleaves insulin-like growth factor-binding proteins (IGFBPs) to release bioactive levels of free insulin-like growth factor. Specific and potent inhibitors of PAPP-A may further elucidate the biological functions of this protease and could prove to be of therapeutic value. Phage display was used to discover fully human antibody inhibitors of PAPP-A activity towards IGFBP4 cleavage. Estimates of the inhibition constants for these antibodies were subsequently determined using a novel continuous assay of PAPP-A protease activity that uses an internally quenched synthetic peptide substrate (DX-1655). DX-1655 was hydrolyzed by PAPP-A with a K(m) of 33 muM and a k(cat) of 0.3 s(-1) (k(cat)/K(m)=9.1x10(3) M(-1) s(-1)). PAPP-A activity towards DX-1655 displays a bell-shaped pH profile, with pK(a) values of 8.2 and 10.8 and a maximum rate at approximately pH 9.5. Using this continuous assay, we measured apparent K(i) values of 1.7+/-0.2 and 7.4+/-1.5 nM for the F2 and D9 antibodies, respectively.


Antibodies/immunology , Antibodies/pharmacology , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/immunology , Amino Acid Sequence , Humans , Hydrogen-Ion Concentration , Hydrolysis/drug effects , Kinetics , Molecular Sequence Data , Peptide Library , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Substrate Specificity
14.
J Cell Physiol ; 204(3): 848-58, 2005 Sep.
Article En | MEDLINE | ID: mdl-15754336

Glucocorticoids (GCs) at physiological concentrations stimulate osteoprogenitor proliferation and differentiation in rat bone cell populations, and this is mediated in part by an increased response to insulin-like growth factors (IGFs). Since IGF binding proteins (IGFBPs) modulate IGF actions, we evaluated whether the increased IGF responsiveness might be associated with decreased inhibitory IGFBP-4 peptide levels. Rat vertebral cells were cultured for up to 20 days with or without dexamethasone (Dex). Cell layer proteins were extracted at day 6, 8, 14, and 20, conditioned media (CM) collected at day 8, 14, and 20, and total RNA isolated at day 14 and 20 of culture. Western blotting showed that cell layer IGFBP-4 levels were lower, while IGFBP-4 protease activity in CM was higher, in Dex-treated cultures. Addition of pregnancy-associated plasma protein-A (PAPP-A) antibody to CM abrogated IGFBP-4 proteolysis. PAPP-A mRNA levels were the same in control and Dex-treated cultures as evaluated by RT-PCR. Our data demonstrate that activity of the IGFBP-4 protease, PAPP-A, in rat bone cell cultures is increased by Dex via post-transcriptional mechanisms. Since IGFBP-4 mRNA levels in Dex-treated cultures were the same as in controls at day 8, slightly lower than in controls at day 14, and higher than in controls at day 20 as shown previously, the decreased IGFBP-4 peptide levels in Dex-treated cultures likely result from increased IGFBP-4 proteolysis by the elevated PAPP-A enzymatic activity. Our findings underscore a novel mechanism whereby GCs increase IGF responses in rat bone cells via PAPP-A-induced IGFBP-4 proteolysis.


Cell Differentiation/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Osteoblasts/drug effects , Pregnancy-Associated Plasma Protein-A/metabolism , Spine/cytology , Stem Cells/drug effects , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Female , Humans , Insulin-Like Growth Factor II/pharmacology , Osteoblasts/cytology , Osteoblasts/metabolism , Pregnancy , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Pregnancy-Associated Plasma Protein-A/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Stem Cells/cytology , Stem Cells/metabolism
15.
Arch Biochem Biophys ; 423(2): 343-50, 2004 Mar 15.
Article En | MEDLINE | ID: mdl-15001398

This study was undertaken to determine the mechanism by which proform of eosinophil major basic protein (proMBP) inhibits the IGFBP-4 proteolytic activity of pregnancy-associated plasma protein (PAPP)-A. Co-overexpression of PAPP-A with proMBP in 293T cells, or co-incubation of 293T cells, respectively, overexpressing proMBP and PAPP-A resulted in the formation of a covalent proMBP-PAPP-A complex and inhibition of IGFBP-4 proteolysis. Similar results were obtained when recombinant proMBP and PAPP-A were incubated in the presence of U2 osteosarcoma cells or when recombinant proMBP was added to the U2 cells overexpressing PAPP-A. In contrast, no formation of covalent proMBP-PAPP-A complex or inhibition of IGFBP-4 proteolysis was observed when recombinant proMBP and PAPP-A were incubated under cell-free conditions, although proMBP was able to interact with PAPP-A in a non-covalent manner. These new findings suggest that formation of covalent proMBP-PAPP-A complex is a cell-mediated event and is required for proMBP to inhibit the catalytic activity of PAPP-A.


Blood Proteins/chemistry , Blood Proteins/metabolism , Enzyme Inhibitors/chemistry , Pregnancy-Associated Plasma Protein-A/chemistry , Pregnancy-Associated Plasma Protein-A/metabolism , Protein Precursors/chemistry , Protein Precursors/physiology , Ribonucleases/chemistry , Ribonucleases/metabolism , Blood Proteins/genetics , Cell Line , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Eosinophil Granule Proteins , Gene Expression , Humans , Immunoblotting , Insulin-Like Growth Factor Binding Protein 4/metabolism , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/genetics , Protein Precursors/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribonucleases/genetics , Transfection
16.
Circulation ; 109(14): 1724-8, 2004 Apr 13.
Article En | MEDLINE | ID: mdl-15023879

BACKGROUND: The metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) has been implicated in coronary plaque disruption. Its endogenous inhibitor, the proform of eosinophil major basic protein (proMBP), may also play a role in this process. Atheromatous plaque disruption often presents as complex angiographic lesions. We sought to assess whether PAPP-A, proMBP, and PAPP-A/ProMBP ratio are markers of angiographic plaque complexity in patients with chronic stable angina. METHODS AND RESULTS: We studied 396 stable angina patients (age 63+/-10 years, 230 men) of whom 289 had angiographically documented coronary artery disease (> or =75% stenosis). All coronary stenoses > or =30% diameter reduction (n =531 in 322 patients) were assessed and classified as complex (n =228) or smooth (n =303) by previously validated criteria. PAPP-A, proMBP, and C-reactive protein (hs-CRP) serum levels were measured by ELISA. Patients with complex coronary stenoses had a significantly (P<0.001) higher PAPP-A/proMBP ratio (3.1+/-1.2 versus 2.7+/-0.8x10(-3)) and PAPP-A levels (5.9+/-1.6 versus 5.1+/-1.4 mIU/L) than those without. On univariate analysis, male gender (P<0.001), age (P<0.001), previous history of myocardial infarction (P=0.013), reduced ejection fraction (P<0.001), severe coronary artery disease (P<0.001), aspirin treatment (P<0.001), PAPP-A levels (P<0.001), and PAPP-A/proMBP ratio (P<0.001) were correlated with the number of complex stenoses. Multiple regression analysis showed that male gender, age, severe coronary artery disease, and PAPP-A/proMBP ratio were independent predictors of the number of angiographically complex stenoses. CONCLUSIONS: In patients with stable angina, PAPP-A and PAPP-A/proMBP ratio are associated with angiographic plaque complexity.


Angina Pectoris/blood , Blood Proteins/physiology , Coronary Artery Disease/blood , Coronary Stenosis/blood , Pregnancy-Associated Plasma Protein-A/physiology , Protein Precursors/physiology , Proteoglycans/physiology , Aged , Angina Pectoris/diagnostic imaging , Angina Pectoris/pathology , Anticoagulants/therapeutic use , Aspirin/therapeutic use , Biomarkers , C-Reactive Protein/analysis , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/pathology , Coronary Stenosis/diagnostic imaging , Coronary Stenosis/pathology , Disease Progression , Eosinophil Major Basic Protein , Female , Humans , Male , Middle Aged , Myocardial Infarction/epidemiology , Predictive Value of Tests , Pregnancy-Associated Plasma Protein-A/analysis , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Protein Precursors/blood , Proteoglycans/blood , Risk Factors , Stroke Volume
17.
Biochem J ; 379(Pt 1): 57-64, 2004 Apr 01.
Article En | MEDLINE | ID: mdl-14705967

PAPP-A (pregnancy-associated plasma protein-A) is produced by hSFs (human skin fibroblasts) and hOBs (human osteoblasts) and enhances the mitogenic activity of IGFs (insulin-like growth factors) by degradation of IGFBP-4 (insulin-like growth factor-binding protein 4). PKC (protein kinase C) activation in these cells led to reduction in IGFBP-4 proteolysis. This study was undertaken to determine the mechanism by which activation of PKC suppresses IGFBP-4 proteolysis. Treatment of hSFs/hOBs with TPA (PMA; 100 nM) reduced IGFBP-4 proteolysis without significantly decreasing the PAPP-A level in the CM (conditioned medium). Immunodepletion of the proform of eosinophil major basic protein (proMBP), a known PAPP-A inhibitor, from CM of TPA-treated cells (TPA CM) failed to increase IGFBP-4 proteolytic activity. Transduction of hSFs with proMBP retrovirus increased the concentration of proMBP up to 30 ng/ml and led to a moderate reduction in IGFBP-4 proteolysis. In contrast, TPA treatment blocked IGFBP-4 proteolysis but failed to induce a detectable amount of proMBP in the CM. While proMBP overexpression led to the formation of a covalent proMBP-PAPP-A complex and reduced the migration of PAPP-A on SDS/PAGE, TPA treatment dose- and time-dependently increased the conversion of a approximately 470 kDa PAPP-A form (PAPP-A470) to a approximately 400 kDa PAPP-A form (PAPP-A400). Since unreduced PAPP-A400 co-migrated with the 400 kDa recombinant PAPP-A homodimer and since PAPP-A monomers from reduced PAPP-A470 and PAPP-A400 co-migrated on SDS/PAGE, conversion of PAPP-A470 to PAPP-A400 is unlikely to be caused by proteolytic cleavage of PAPP-A. Consistent with the data showing that the increase in the ratio of PAPP-A400/PAPP-A470 is correlated with the extent of reduction in IGFBP-4 proteolysis, partially purified PAPP-A400 exhibited a 4-fold reduction in IGFBP-4 proteolytic activity compared with PAPP-A470. These data suggest that a novel mechanism, namely conversion of PAPP-A470 to the less-active PAPP-A400, could account for the TPA-induced suppression of PAPP-A activity.


Fibroblasts/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Osteoblasts/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Pregnancy-Associated Plasma Protein-A/physiology , Protein Kinase C/metabolism , Adult , Antibodies, Monoclonal/pharmacology , Blood Proteins/immunology , Blood Proteins/metabolism , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Culture Media, Conditioned/pharmacology , Dimerization , Enzyme Activation , Eosinophil Granule Proteins , Fibroblasts/drug effects , Humans , Male , Molecular Weight , Osteoblasts/drug effects , Phorbol Esters/pharmacology , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/chemistry , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Precursors/immunology , Protein Precursors/metabolism , Recombinant Fusion Proteins/physiology , Ribonucleases/immunology , Ribonucleases/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transduction, Genetic
18.
J Biol Chem ; 278(17): 14640-6, 2003 Apr 25.
Article En | MEDLINE | ID: mdl-12571229

Bikunin, a Kunitz-type protease inhibitor, could potentially suppress tumor cell invasion and metastasis. Our previous study revealed that overexpression of bikunin in a human ovarian cancer cell line, HRA, resulted in a down-regulation in uPA and uPAR gene expression. For identifying the full repertoire of bikunin-regulated genes, a cDNA microarray hybridization screening was conducted using mRNA from bikunin-treated or bikunin-transfected HRA cells. A number of bikunin-regulated genes were identified, and their regulation was confirmed by Northern blot analysis. Our screen identified 11 bikunin-stimulated genes and 29 bikunin-repressed genes. The identified genes can indeed be classified into distinct subsets. These include transcriptional regulators, oncogenes/tumor suppressor genes, signaling molecules, growth/cell cycle, invasion/metastasis, cytokines, apoptosis, ion channels, extracellular matrix proteins, as well as some proteases. This screen identified suppression of several genes such as CDC-like kinase, LIM domain binding, Ets domain transcription factor, Rho GTPase-activating protein, tyrosine phosphorylation-regulated kinase, hyaluronan-binding protein, matriptase, and pregnancy-associated plasma protein-A (PAPP-A), which have previously been implicated in enhancing tumor promotion. Northern blot analysis confirmed that several genes including matriptase and PAPP-A were down-regulated by bikunin by approximately 9-fold. Further, genetic inhibition of matriptase or PAPP-A could lead to diminished invasion. These results show that bikunin alters the pattern of gene expression in HRA cells leading to a block in cell invasion.


Membrane Glycoproteins/physiology , Ovarian Neoplasms/pathology , Trypsin Inhibitor, Kunitz Soybean , Cell Movement/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Glycoproteins/pharmacology , Oligonucleotide Array Sequence Analysis , Ovarian Neoplasms/genetics , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Serine Endopeptidases/drug effects , Serine Proteinase Inhibitors/genetics , Serine Proteinase Inhibitors/pharmacology , Serine Proteinase Inhibitors/physiology , Transfection , Trypsin/drug effects , Tumor Cells, Cultured
19.
Biol Reprod ; 68(1): 77-86, 2003 Jan.
Article En | MEDLINE | ID: mdl-12493698

In mammalian ovaries, terminal follicular growth is accompanied by a decrease in levels of intrafollicular insulin-like growth factor binding protein-2 (IGFBP-2) and IGFBP-4. The decrease in IGFBP-4 is essentially due to an increase in proteolytic degradation by intrafollicular pregnancy-associated plasma protein-A (PAPP-A) in growing healthy follicles. In contrast, the decrease in IGFBP-2 is partly due to a decrease in mRNA expression by follicular cells and also to an increase in IGFBP-2 proteolytic degradation, as previously shown in ewes and sows. In the present work we show that bovine and porcine preovulatory follicular fluid contains a proteolytic activity that degrades IGFBP-2. Bovine and porcine preovulatory follicular fluids contain undetectable levels of native IGFBP-2 as assessed by Western ligand blotting in comparison with the corresponding serum. In contrast, much higher levels of 23- and 12-kDa proteolytic fragments were found by immunoblotting in bovine and porcine preovulatory follicular fluid than in the corresponding serum. Moreover, bovine and porcine preovulatory follicular fluids were able to induce proteolytic degradation of exogenous IGFBP-2, and this degradation was enhanced by insulin-like growth factors. Intrafollicular IGFBP-2 proteolytic activity was surprisingly immunoneutralized in both species by a polyclonal antibody raised against human PAPP-A. In addition, recombinant human PAPP-A (rhPAPP-A) was able to cleave IGFBP-2 between Gln165 and Met166 in vitro, generating 23- and 12-kDa proteolytic fragments. IGFBP-2 was shown to be less sensitive than IGFBP-4 to cleavage by rhPAPP-A in vitro. As in follicular fluid, cleavage of IGFBP-2 by rhPAPP-A was dose-dependently enhanced by IGFs and inhibited by a peptide derived from the heparin-binding domain of IGFBP-5 (P5). Finally, Biacore analysis showed that P5 peptide-induced inhibition of IGFBP-2 cleavage was due to a direct interaction of P5 with PAPP-A rather than with IGFBP-2. Overall, these data show that in bovine and porcine preovulatory follicles, PAPP-A is responsible for IGF-dependent IGFBP-2 degradation. During follicular growth, the increase in IGFBP-2 cleavage by PAPP-A, as well as the decrease in IGFBP-2 expression, are responsible for the decrease in intact IGFBP-2 levels and the increase in IGF bioavailability. In atretic follicles, the increase and decrease in IGFBP-2 and PAPP-A mRNA expression, respectively, as well as the inhibition of PAPP-A activity by heparin-binding domains present in IGFBP-5 or other proteins, might participate in higher IGFBP-2 levels and a decrease in IGF bioavailability.


Insulin-Like Growth Factor Binding Protein 2/metabolism , Ovarian Follicle/metabolism , Pregnancy-Associated Plasma Protein-A/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cattle , Female , Follicular Fluid/metabolism , Follicular Phase/metabolism , Humans , In Vitro Techniques , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor Binding Protein 4/metabolism , Insulin-Like Growth Factor Binding Protein 5/genetics , Insulin-Like Growth Factor Binding Protein 5/metabolism , Kinetics , Molecular Sequence Data , Neutralization Tests , Peptide Fragments/genetics , Peptide Fragments/metabolism , Pregnancy , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/metabolism , Somatomedins/metabolism , Sus scrofa
20.
J Biol Chem ; 275(40): 31128-33, 2000 Oct 06.
Article En | MEDLINE | ID: mdl-10913121

Pregnancy-associated plasma protein-A (PAPP-A), originally known from human pregnancy serum, has recently been demonstrated to be a metzincin superfamily metalloproteinase involved in normal and pathological insulin-like growth factor (IGF) physiology. PAPP-A specifically cleaves IGF-binding protein (IGFBP)-4, one of six antagonists of IGF action, which results in release of IGF bound to IGFBP-4. IGFBP-4 is the only known PAPP-A substrate. Its cleavage by PAPP-A uniquely depends on the presence of IGF. We here report mammalian expression and purification of recombinant 1547-residue PAPP-A (rPAPP-A). The recombinant protein is secreted as a homodimer of about 400 kDa composed of two 200-kDa disulfide-bound subunits. Antigenically and functionally, rPAPP-A behaves like the native protein. In human pregnancy, PAPP-A is known to circulate as a 500-kDa disulfide-bound 2:2 complex with the proform of eosinophil major basic protein (proMBP), PAPP-A/proMBP. A comparison between rPAPP-A and pregnancy serum PAPP-A/proMBP complex surprisingly reveals a difference greater than 100-fold in proteolytic activity, showing that proMBP functions as a proteinase inhibitor in vivo. We find that polyclonal antibodies against PAPP-A abrogate all detectable IGFBP-4 proteolytic activity in pregnancy serum, pointing at PAPP-A as the dominating, if not the only, IGFBP-4 proteinase present in the circulation. We further show that pregnancy serum and plasma contain traces (<1%) of uncomplexed PAPP-A with a much higher specific activity than the PAPP-A/proMBP complex. The measurable activity of the PAPP-A/proMBP complex probably results from the presence of a minor subpopulation of partly inhibited PAPP-A that exists in a 2:1 complex with proMBP. Inhibition of PAPP-A by proMBP represents a novel inhibitory mechanism with the enzyme irreversibly bound to its inhibitor by disulfide bonds.


Blood Proteins/biosynthesis , Blood Proteins/metabolism , Recombinant Proteins/metabolism , Ribonucleases , Blood Proteins/chemistry , Blotting, Western , Cell Line , Chromatography, Ion Exchange , DNA, Complementary/metabolism , Disulfides , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors , Enzyme-Linked Immunosorbent Assay , Eosinophil Granule Proteins , Eosinophils/chemistry , Female , Humans , Insulin-Like Growth Factor Binding Protein 4/blood , Metalloendopeptidases/biosynthesis , Metalloendopeptidases/chemistry , Plasmids/metabolism , Pregnancy , Pregnancy-Associated Plasma Protein-A/antagonists & inhibitors , Pregnancy-Associated Plasma Protein-A/metabolism , Pregnancy-Associated Plasma Protein-A/physiology , Somatomedins/metabolism , Transfection
...