Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Metabolism ; 132: 155218, 2022 07.
Article En | MEDLINE | ID: mdl-35588861

BACKGROUND: Diabetic nephropathy (DN) is a serious complication of diabetes and a common cause of end stage renal failure. Insulin-like growth factor (IGF)-signaling has been implicated in DN, but is mechanistically poorly understood. Here, we assessed the activity of the metalloproteinase PAPP-A, an activator of IGF activity, and its possible interaction with the endogenous PAPP-A inhibitors stanniocalcin (STC)-1 and -2 in the mammalian kidney under normal and hyperglycemic conditions. METHODS AND RESULTS: Immunohistochemistry demonstrated that PAPP-A, its proteolytic substrate IGF binding protein-4, STC1 and STC2 are present in the human kidney. Endogenous inhibited complexes of PAPP-A (PAPP-A:STC1 and PAPP-A:STC2) were demonstrated in media conditioned by human mesangial cells (HMCs), suggesting that PAPP-A activity is regulated by the STCs in kidney tissue. A method for the selective detection of active PAPP-A in tissue was developed and a significant increase in glomerular active PAPP-A in human diabetic kidney relative to normal was observed. In DN patients, the estimated glomerular filtration rate correlated with PAPP-A activity. In diabetic mice, glomerular growth was reduced when PAPP-A activity was antagonized by adeno-associated virus-mediated overexpression of STC2. CONCLUSION: We propose that PAPP-A activity in renal tissue is precisely balanced by STC1 and STC2. An imbalance in this equilibrium causing increased PAPP-A enzymatic activity potentially contributes to the development of DN, and thus, therapeutic targeting of PAPP-A activity may represent a novel strategy for its treatment.


Diabetes Mellitus, Experimental , Diabetic Nephropathies , Pregnancy-Associated Plasma Protein-A , Animals , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Humans , Hypertrophy , Intercellular Signaling Peptides and Proteins/metabolism , Mammals/metabolism , Mice , Pregnancy-Associated Plasma Protein-A/metabolism , Proteolysis
2.
J Biol Chem ; 294(45): 16684-16697, 2019 11 08.
Article En | MEDLINE | ID: mdl-31543504

Macrophage phagocytosis is required for effective clearance of invading bacteria and other microbes. Coordinated phosphoinositide signaling is critical both for phagocytic particle engulfment and subsequent phagosomal maturation to a degradative organelle. Phosphatidylinositol 3-phosphate (PtdIns(3)P) is a phosphoinositide that is rapidly synthesized and degraded on phagosomal membranes, where it recruits FYVE domain- and PX motif-containing proteins that promote phagosomal maturation. However, the molecular mechanisms that regulate PtdIns(3)P removal from the phagosome have remained unclear. We report here that a myotubularin PtdIns(3)P 3-phosphatase, myotubularin-related protein-4 (MTMR4), regulates macrophage phagocytosis. MTMR4 overexpression reduced and siRNA-mediated Mtmr4 silencing increased levels of cell-surface immunoglobulin receptors (i.e. Fcγ receptors (FcγRs)) on RAW 264.7 macrophages, associated with altered pseudopodal F-actin. Furthermore, MTMR4 negatively regulated the phagocytosis of IgG-opsonized particles, indicating that MTMR4 inhibits FcγR-mediated phagocytosis, and was dynamically recruited to phagosomes of macrophages during phagocytosis. MTMR4 overexpression decreased and Mtmr4-specific siRNA expression increased the duration of PtdIns(3)P on phagosomal membranes. Macrophages treated with Mtmr4-specific siRNA were more resistant to Mycobacterium marinum-induced phagosome arrest, associated with increased maturation of mycobacterial phagosomes, indicating that extended PtdIns(3)P signaling on phagosomes in the Mtmr4-knockdown cells permitted trafficking of phagosomes to acidic late endosomal and lysosomal compartments. In conclusion, our findings indicate that MTMR4 regulates PtdIns(3)P degradation in macrophages and thereby controls phagocytosis and phagosomal maturation.


Phagocytosis , Phagosomes/metabolism , Phosphatidylinositol Phosphates/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Actins/metabolism , Animals , Endosomes/metabolism , Humans , Immunoglobulin G/immunology , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , Mycobacterium marinum/pathogenicity , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RAW 264.7 Cells , RNA Interference , RNA, Small Interfering/metabolism , Receptors, IgG/metabolism , Signal Transduction
3.
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
4.
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
5.
J Biol Chem ; 290(6): 3430-9, 2015 Feb 06.
Article En | MEDLINE | ID: mdl-25533459

Mammalian stanniocalcin-2 (STC2) is a secreted polypeptide widely expressed in developing and adult tissues. However, although transgenic expression in mice is known to cause severe dwarfism, and targeted deletion of STC2 causes increased postnatal growth, its precise biological role is still unknown. We found that STC2 potently inhibits the proteolytic activity of the growth-promoting metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A). Proteolytic inhibition requires covalent binding of STC2 to PAPP-A and is mediated by a disulfide bond, which involves Cys-120 of STC2. Binding of STC2 prevents PAPP-A cleavage of insulin-like growth factor-binding protein (IGFBP)-4 and hence release within tissues of bioactive IGF, required for normal growth. Concordantly, we show that STC2 efficiently inhibits PAPP-A-mediated IGF receptor signaling in vitro and that transgenic mice expressing a mutated variant of STC2, STC2(C120A), which is unable to inhibit PAPP-A, grow like wild-type mice. Our work identifies STC2 as a novel proteinase inhibitor and a previously unrecognized extracellular component of the IGF system.


Glycoproteins/metabolism , Growth/genetics , Pregnancy-Associated Plasma Protein-A/metabolism , Proteolysis , Amino Acid Sequence , Animals , Cells, Cultured , Cysteine/chemistry , Cysteine/genetics , Female , Glycoproteins/chemistry , Glycoproteins/genetics , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 4/metabolism , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Male , Mice , Molecular Sequence Data , Mutation , Protein Binding , Receptor, IGF Type 1/metabolism
6.
J Cell Sci ; 127(Pt 23): 5027-37, 2014 Dec 01.
Article En | MEDLINE | ID: mdl-25236600

Pregnancy-associated plasma protein A2 (PAPP-A2, also known as pappalysin-2) is a large metalloproteinase that is known to be required for normal postnatal growth and bone development in mice. We here report the detection of zebrafish papp-a2 mRNA in the chordamesoderm, notochord and lower jaw of zebrafish (Danio rerio) embryos, and that papp-a2-knockdown embryos display broadened axial mesoderm, notochord bends and severely reduced cranial cartilages. Genetic data link these phenotypes to insulin-like growth factor (Igf)-binding protein-3 (Igfbp-3) and bone morphogenetic protein (Bmp) signaling, and biochemical analysis show specific Igfbp-3 proteolysis by Papp-a2, implicating Papp-a2 in the modulation of Bmp signaling by Igfbp-3 proteolysis. Knockdown of papp-a2 additionally resulted in angiogenesis defects, strikingly similar to previous observations in embryos with mutations in components of the Notch system. Accordingly, we find that Notch signaling is modulated by Papp-a2 in vivo, and, furthermore, that human PAPP-A2 is capable of modulating Notch signaling independently of its proteolytic activity in cell culture. Based on these results, we conclude that Papp-a2 modulates Bmp and Notch signaling by independent mechanisms in zebrafish embryos. In conclusion, these data link pappalysin function in zebrafish to two different signaling pathways outside the IGF system.


Cartilage/enzymology , Neovascularization, Physiologic , Pregnancy-Associated Plasma Protein-A/metabolism , Skull/enzymology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Amino Acid Sequence , Animals , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cartilage/embryology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Genotype , HEK293 Cells , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Insulin-Like Growth Factor Binding Protein 4/metabolism , Molecular Sequence Data , Phenotype , Pregnancy-Associated Plasma Protein-A/genetics , RNA, Messenger/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Skull/embryology , Time Factors , Transfection , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
...