1
|
Petersen JF, Tiittanen V, Wittfooth S, Løkkegaard E, Friis-Hansen LJ. Exploring free pregnancy associated plasma protein a (fPAPP-A) as a biomarker in early pregnancy. Pract Lab Med 2024; 42:e00428. [PMID: 39411186 PMCID: PMC11474183 DOI: 10.1016/j.plabm.2024.e00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/16/2024] [Accepted: 09/15/2024] [Indexed: 10/19/2024] Open
Abstract
Objectives In combined first trimester screening for Down syndrome, Pregnancy-Associated Plasma Protein A (PAPP-A) is pivotal. PAPP-A tests evaluate total PAPP-A, consisting of the biologically active free PAPP-A (fPAPP-A) and PAPP-A complexed with eosinophil major basic protein's proform (proMBP). While PAPP-A is well-researched, limited understanding persists regarding fPAPP-A's first trimester concentrations and diagnostic utility. Design and methods: PAPP-A and fPAPP-A levels were gauged in 602 serum samples at 2-week intervals (gestational weeks 4-14) from 159 women with delivery of a healthy neonate and 80 samples from 37 miscarriages. The final sample at the time of diagnosis from women who miscarried was included in analyses. Results During the first trimester, PAPP-A and fPAPP-A levels displayed significant and strong correlation (r = 0.94), with median values doubling weekly. Free PAPP-A constituted only 3.0 % of PAPP-A over gestational weeks. Low fPAPP-A linked to miscarriage (p < 0.001), maternal weight (p < 0.001), and smoking (p = 0.02). For miscarriage prediction fPAPP-A was equal to PAPP-A (area under the receiver operating characteristics curve 0.79 vs. 0.81, p = 0.44). Conclusions Investigating fPAPP-A presence and concentration directly in first trimester serum has not been done previously. This study report lower fPAPP-A values than anticipated from prior enzymatic studies of fPAPP-A. fPAPP-A was not superior to PAPP-A as a first trimester biomarker in this dataset.
Collapse
Affiliation(s)
- Jesper Friis Petersen
- Department of Obstetrics and Gynecology, North Zealand Hospital, Dyrehavevej 29, 3400, Hillerød, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Vilma Tiittanen
- Biotechnology Unit, Department of Life Technologies, 20014, University of Turku, Finland
| | - Saara Wittfooth
- Biotechnology Unit, Department of Life Technologies, 20014, University of Turku, Finland
| | - Ellen Løkkegaard
- Department of Obstetrics and Gynecology, North Zealand Hospital, Dyrehavevej 29, 3400, Hillerød, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Lennart Jan Friis-Hansen
- Department of Clinical Biochemistry, Bispebjerg Hospital University Hospital, Nielsine Nielsens Vej 2, 2400, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
2
|
Güemes M, Martín-Rivada Á, Corredor B, Enes P, Canelles S, Barrios V, Argente J. Implication of Pappalysins and Stanniocalcins in the Bioavailability of IGF-I in Children With Type 1 Diabetes Mellitus. J Endocr Soc 2024; 8:bvae081. [PMID: 38712328 PMCID: PMC11071684 DOI: 10.1210/jendso/bvae081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Indexed: 05/08/2024] Open
Abstract
Context Anomalies in the growth hormone (GH)/insulin-like growth factor (IGF) axis, are common in children with type 1 diabetes mellitus (T1DM), even in those reaching a normal or near-normal final height. However, concentrations of the IGF bioavailability regulatory factors (pappalysins [PAPP-As] and stanniocalcins [STCs]) have not been reported in children with T1DM. Objective To determine serum concentrations of PAPP-As and STCs in children at diagnosis of T1DM and after insulin treatment and the correlation of these factors with other members of the GH/IGF axis, beta-cell insulin reserve, auxology, and nutritional status. Methods A single-center prospective observational study including 47 patients (59.5% male), with T1DM onset at median age of 9.2 years (interquartile range: 6.3, 11.9) was performed. Blood and anthropometric data were collected at diagnosis and after 6 and 12 months of treatment. Results At 6 and 12 months after T1DM diagnosis, there was improvement in the metabolic control (decrease in glycated hemoglobin [HbA1c] at 12 months -3.66 [95% CI: -4.81, -2.05], P = .001), as well as in body mass index SD and height SD (not statistically significant). STC2 increased (P < .001) and PAPP-A2 decreased (P < .001) at 6 and 12 months of treatment onset (P < .001), which was concurrent with increased total IGF-I and IGF-binding protein concentrations, with no significant modification in free IGF-I concentrations. HbA1c correlated with PAPP-A2 (r = +0.41; P < .05) and STC2 (r = -0.32; P < .05). Conclusion Implementation of insulin treatment after T1DM onset modifies various components of the circulating IGF system, including PAPP-A2 and STC2. How these modifications modulate linear growth remains unknown.
Collapse
Affiliation(s)
- María Güemes
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
| | - Álvaro Martín-Rivada
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
| | - Beatriz Corredor
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
| | - Patricia Enes
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
| | - Sandra Canelles
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
| | - Vicente Barrios
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Jesús Argente
- Department of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute La Princesa, 28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, 28029 Madrid, Spain
- IMDEA, Food Institute, CEIUAM+CSI, 28049 Madrid, Spain
| |
Collapse
|
3
|
Poddar A, Ahmady F, Rao SR, Sharma R, Kannourakis G, Prithviraj P, Jayachandran A. The role of pregnancy associated plasma protein-A in triple negative breast cancer: a promising target for achieving clinical benefits. J Biomed Sci 2024; 31:23. [PMID: 38395880 PMCID: PMC10885503 DOI: 10.1186/s12929-024-01012-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Pregnancy associated plasma protein-A (PAPP-A) plays an integral role in breast cancer (BC), especially triple negative breast cancer (TNBC). This subtype accounts for the most aggressive BC, possesses high tumor heterogeneity, is least responsive to standard treatments and has the poorest clinical outcomes. There is a critical need to address the lack of effective targeted therapeutic options available. PAPP-A is a protein that is highly elevated during pregnancy. Frequently, higher PAPP-A expression is detected in tumors than in healthy tissues. The increase in expression coincides with increased rates of aggressive cancers. In BC, PAPP-A has been demonstrated to play a role in tumor initiation, progression, metastasis including epithelial-mesenchymal transition (EMT), as well as acting as a biomarker for predicting patient outcomes. In this review, we present the role of PAPP-A, with specific focus on TNBC. The structure and function of PAPP-A, belonging to the pappalysin subfamily, and its proteolytic activity are assessed. We highlight the link of BC and PAPP-A with respect to the IGFBP/IGF axis, EMT, the window of susceptibility and the impact of pregnancy. Importantly, the relevance of PAPP-A as a TNBC clinical marker is reviewed and its influence on immune-related pathways are explored. The relationship and mechanisms involving PAPP-A reveal the potential for more treatment options that can lead to successful immunotherapeutic targets and the ability to assist with better predicting clinical outcomes in TNBC.
Collapse
Affiliation(s)
- Arpita Poddar
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
- RMIT University, Victoria, Australia
| | - Farah Ahmady
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Sushma R Rao
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Revati Sharma
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Prashanth Prithviraj
- Fiona Elsey Cancer Research Institute, Victoria, Australia
- Federation University, Victoria, Australia
| | - Aparna Jayachandran
- Fiona Elsey Cancer Research Institute, Victoria, Australia.
- Federation University, Victoria, Australia.
| |
Collapse
|
4
|
Barrios V, Martín-Rivada Á, Guerra-Cantera S, Campillo-Calatayud A, Camarneiro RA, Graell M, Chowen JA, Argente J. Reduction in Pappalysin-2 Levels and Lower IGF-I Bioavailability in Female Adolescents With Anorexia Nervosa. J Clin Endocrinol Metab 2024; 109:e920-e931. [PMID: 38066647 DOI: 10.1210/clinem/dgad713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Indexed: 02/21/2024]
Abstract
CONTEXT Anorexia nervosa (AN) can cause severe undernutrition associated with alterations in the IGF axis. Pappalysins (PAPP-A, PAPP-A2) and stanniocalcins (STC-1, STC-2) modulate IGF binding-protein (IGFBP) cleavage and IGF bioavailability, but their implications in AN are unknown. OBJECTIVE We determined serum levels of PAPP-As and STCs in relationship with classical IGF axis parameters in female adolescents with AN and their association with nutritional status and secondary amenorrhea. METHODS Parameters of the IGF axis were determined in fasting serum samples of 68 female adolescents with AN at diagnosis and 62 sex- and age-matched controls. Standardized body mass index (BMI) and bone mineral density (BMD) were calculated. RESULTS Patients with AN had lower concentrations of total and free IGF-I, total IGFBP-3, acid-labile subunit (ALS), insulin, PAPP-A2, STC-1, and STC-2 and higher levels of IGF-II and IGFBP-2. Their free/total IGF-I ratio was decreased and the intact/total IGFBP-3 and -4 ratios increased. BMI was directly related to total IGF-I and intact IGFBP-3 and inversely with IGFBP-2 and intact IGFBP-4. Weight loss was directly correlated with intact IGFBP-4 and negatively with intact IGFBP-3, ALS, STC-2, and PAPP-A2 concentrations. BMD was directly related to intact IGFBP-3 and inversely with intact IGFBP-4 and PAPP-A2 levels. Patients with amenorrhea had lower levels of total IGF-I and IGFBP-3 than those with menses. CONCLUSION The reduction of PAPP-A2 in patients with AN may be involved in a decline in IGFBP cleavage, which could underlie the decrease in IGF-I bioavailability that is influenced by nutritional status and amenorrhea.
Collapse
Affiliation(s)
- Vicente Barrios
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn, Instituto de Salud Carlos III, E-28009 Madrid, Spain
| | - Álvaro Martín-Rivada
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
| | - Santiago Guerra-Cantera
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
| | - Ana Campillo-Calatayud
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
| | - Ricardo A Camarneiro
- Department of Psychiatry and Clinical Psychology, Hospital Infantil Universitario Niño Jesús, E-28009 Madrid, Spain
| | - Montserrat Graell
- Department of Psychiatry and Clinical Psychology, Hospital Infantil Universitario Niño Jesús, E-28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, E-28009 Madrid, Spain
| | - Julie A Chowen
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn, Instituto de Salud Carlos III, E-28009 Madrid, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, E-28049 Madrid, Spain
| | - Jesús Argente
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", E-28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn, Instituto de Salud Carlos III, E-28009 Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, E-28009 Madrid, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, E-28049 Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| |
Collapse
|
5
|
Conover CA, Oxvig C. The Pregnancy-Associated Plasma Protein-A (PAPP-A) Story. Endocr Rev 2023; 44:1012-1028. [PMID: 37267421 DOI: 10.1210/endrev/bnad017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/01/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) was first identified in the early 1970s as a placental protein of unknown function, present at high concentrations in the circulation of pregnant women. In the mid-to-late 1990s, PAPP-A was discovered to be a metzincin metalloproteinase, expressed by many nonplacental cells, that regulates local insulin-like growth factor (IGF) activity through cleavage of high-affinity IGF binding proteins (IGFBPs), in particular IGFBP-4. With PAPP-A as a cell surface-associated enzyme, the reduced affinity of the cleavage fragments results in increased IGF available to bind and activate IGF receptors in the pericellular environment. This proteolytic regulation of IGF activity is important, since the IGFs promote proliferation, differentiation, migration, and survival in various normal and cancer cells. Thus, there has been a steady growth in investigation of PAPP-A structure and function outside of pregnancy. This review provides historical perspective on the discovery of PAPP-A and its structure and cellular function, highlights key studies of the first 50 years in PAPP-A research, and introduces new findings from recent years.
Collapse
Affiliation(s)
- Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
6
|
Lionikas A, Hernandez Cordero AI, Kilikevicius A, Carroll AM, Bewick GS, Bunger L, Ratkevicius A, Heisler LK, Harboe M, Oxvig C. Stanniocalcin-2 inhibits skeletal muscle growth and is upregulated in functional overload-induced hypertrophy. Physiol Rep 2023; 11:e15793. [PMID: 37568262 PMCID: PMC10510475 DOI: 10.14814/phy2.15793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
AIMS Stanniocalcin-2 (STC2) has recently been implicated in human muscle mass variability by genetic analysis. Biochemically, STC2 inhibits the proteolytic activity of the metalloproteinase PAPP-A, which promotes muscle growth by upregulating the insulin-like growth factor (IGF) axis. The aim was to examine if STC2 affects skeletal muscle mass and to assess how the IGF axis mediates muscle hypertrophy induced by functional overload. METHODS We compared muscle mass and muscle fiber morphology between Stc2-/- (n = 21) and wild-type (n = 15) mice. We then quantified IGF1, IGF2, IGF binding proteins -4 and -5 (IGFBP-4, IGFBP-5), PAPP-A and STC2 in plantaris muscles of wild-type mice subjected to 4-week unilateral overload (n = 14). RESULTS Stc2-/- mice showed up to 10% larger muscle mass compared with wild-type mice. This increase was mediated by greater cross-sectional area of muscle fibers. Overload increased plantaris mass and components of the IGF axis, including quantities of IGF1 (by 2.41-fold, p = 0.0117), IGF2 (1.70-fold, p = 0.0461), IGFBP-4 (1.48-fold, p = 0.0268), PAPP-A (1.30-fold, p = 0.0154) and STC2 (1.28-fold, p = 0.019). CONCLUSION Here we provide evidence that STC2 is an inhibitor of muscle growth upregulated, along with other components of the IGF axis, during overload-induced muscle hypertrophy.
Collapse
Affiliation(s)
- Arimantas Lionikas
- School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Ana I. Hernandez Cordero
- Centre for Heart Lung InnovationUniversity of British Columbia, St. Paul's HospitalVancouverCanada
| | - Audrius Kilikevicius
- Department of Health Promotion and RehabilitationLithuanian Sports UniversityKaunasLithuania
| | - Andrew M. Carroll
- The New Zealand Institute for Plant & Food Research LimitedPalmerston NorthNew Zealand
| | - Guy S. Bewick
- School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Lutz Bunger
- Animal Genetics Company (AnGeCo)EdinburghScotland
| | - Aivaras Ratkevicius
- Department of Health Promotion and RehabilitationLithuanian Sports UniversityKaunasLithuania
| | - Lora K. Heisler
- School of Medicine, Medical Sciences and NutritionUniversity of AberdeenAberdeenUK
| | - Mette Harboe
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| | - Claus Oxvig
- Department of Molecular Biology and GeneticsAarhus UniversityAarhusDenmark
| |
Collapse
|
7
|
Oxvig C, Conover CA. The Stanniocalcin-PAPP-A-IGFBP-IGF Axis. J Clin Endocrinol Metab 2023; 108:1624-1633. [PMID: 36718521 DOI: 10.1210/clinem/dgad053] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 02/01/2023]
Abstract
The pappalysin metalloproteinases, PAPP-A and PAPP-A2, have emerged as highly specific proteolytic enzymes involved in the regulation of insulin-like growth factor (IGF) signaling. The only known pappalysin substrates are a subset of the IGF binding proteins (IGFBPs), which bind IGF-I or IGF-II with high affinity to antagonize receptor binding. Thus, by cleaving IGFBPs, the pappalysins have the potential to increase IGF bioactivity and hence promote IGF signaling. This is relevant both in systemic and local IGF regulation, in normal and several pathophysiological conditions. Stanniocalcin-1 and -2 were recently found to be potent pappalysin inhibitors, thus comprising the missing components of a complete proteolytic system, the stanniocalcin-PAPP-A-IGFBP-IGF axis. Here, we provide the biological context necessary for understanding the properties of this molecular network, and we review biochemical data, animal experiments, clinical data, and genetic data supporting the physiological operation of this branch as an important part of the IGF system. However, although in vivo data clearly illustrate its power, it is a challenge to understand its subtle operation, for example, multiple equilibria and inhibitory kinetics may determine how, where, and when the IGF receptor is stimulated. In addition, literally all of the regulatory proteins have suspected or known activities that are not directly related to IGF signaling. How such activities may integrate with IGF signaling is also important to address in the future.
Collapse
Affiliation(s)
- Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000 C, Aarhus, Denmark
| | - Cheryl A Conover
- Division of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Zhong Q, Chu H, Wang G, Zhang C, Li R, Guo F, Meng X, Lei X, Zhou Y, Ren R, Tao L, Li N, Gao N, Wei Y, Qiao J, Hang J. Structural insights into the covalent regulation of PAPP-A activity by proMBP and STC2. Cell Discov 2022; 8:137. [PMID: 36550107 PMCID: PMC9780223 DOI: 10.1038/s41421-022-00502-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 11/25/2022] [Indexed: 12/24/2022] Open
Abstract
Originally discovered in the circulation of pregnant women as a protein secreted by placental trophoblasts, the metalloprotease pregnancy-associated plasma protein A (PAPP-A) is also widely expressed by many other tissues. It cleaves insulin-like growth factor-binding proteins (IGFBPs) to increase the bioavailability of IGFs and plays essential roles in multiple growth-promoting processes. While the vast majority of the circulatory PAPP-A in pregnancy is proteolytically inactive due to covalent inhibition by proform of eosinophil major basic protein (proMBP), the activity of PAPP-A can also be covalently inhibited by another less characterized modulator, stanniocalcin-2 (STC2). However, the structural basis of PAPP-A proteolysis and the mechanistic differences between these two modulators are poorly understood. Here we present two cryo-EM structures of endogenous purified PAPP-A in complex with either proMBP or STC2. Both modulators form 2:2 heterotetramer with PAPP-A and establish extensive interactions with multiple domains of PAPP-A that are distal to the catalytic cleft. This exosite-binding property results in a steric hindrance to prevent the binding and cleavage of IGFBPs, while the IGFBP linker region-derived peptides harboring the cleavage sites are no longer sensitive to the modulator treatment. Functional investigation into proMBP-mediated PAPP-A regulation in selective intrauterine growth restriction (sIUGR) pregnancy elucidates that PAPP-A and proMBP collaboratively regulate extravillous trophoblast invasion and the consequent fetal growth. Collectively, our work reveals a novel covalent exosite-competitive inhibition mechanism of PAPP-A and its regulatory effect on placental function.
Collapse
Affiliation(s)
- Qihang Zhong
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Honglei Chu
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Guopeng Wang
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Cheng Zhang
- grid.412474.00000 0001 0027 0586Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Rong Li
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Fusheng Guo
- grid.11135.370000 0001 2256 9319Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xinlu Meng
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Xiaoguang Lei
- grid.11135.370000 0001 2256 9319Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Department of Chemical Biology, College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China ,grid.510951.90000 0004 7775 6738Institute for Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong China
| | - Youli Zhou
- grid.10784.3a0000 0004 1937 0482School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong China
| | - Ruobing Ren
- grid.10784.3a0000 0004 1937 0482School of Life and Health Sciences, The Chinese University of Hong Kong, Shenzhen, Guangdong China ,grid.8547.e0000 0001 0125 2443Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Lin Tao
- grid.412636.40000 0004 1757 9485Department of Orthopedics, First Hospital of China Medical University, Shenyang, Liaoning China
| | - Ningning Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China
| | - Ning Gao
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, School of Life Sciences, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yuan Wei
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| | - Jie Qiao
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China ,grid.11135.370000 0001 2256 9319Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jing Hang
- grid.411642.40000 0004 0605 3760Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China ,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, China ,grid.411642.40000 0004 0605 3760National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
| |
Collapse
|
9
|
Structure of the proteolytic enzyme PAPP-A with the endogenous inhibitor stanniocalcin-2 reveals its inhibitory mechanism. Nat Commun 2022; 13:6084. [PMID: 36257932 PMCID: PMC9579167 DOI: 10.1038/s41467-022-33698-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/27/2022] [Indexed: 12/24/2022] Open
Abstract
The metzincin metalloproteinase PAPP-A plays a key role in the regulation of insulin-like growth factor (IGF) signaling by specific cleavage of inhibitory IGF binding proteins (IGFBPs). Using single-particle cryo-electron microscopy (cryo-EM), we here report the structure of PAPP-A in complex with its endogenous inhibitor, stanniocalcin-2 (STC2), neither of which have been reported before. The highest resolution (3.1 Å) was obtained for the STC2 subunit and the N-terminal approximately 1000 residues of the PAPP-A subunit. The 500 kDa 2:2 PAPP-A·STC2 complex is a flexible multidomain ensemble with numerous interdomain contacts. In particular, a specific disulfide bond between the subunits of STC2 and PAPP-A prevents dissociation, and interactions between STC2 and a module located in the very C-terminal end of the PAPP-A subunit prevent binding of its main substrate, IGFBP-4. While devoid of activity towards IGFBP-4, the active site cleft of the catalytic domain is accessible in the inhibited PAPP-A·STC2 complex, as shown by its ability to hydrolyze a synthetic peptide derived from IGFBP-4. Relevant to multiple human pathologies, this unusual mechanism of proteolytic inhibition may support the development of specific pharmaceutical agents, by which IGF signaling can be indirectly modulated.
Collapse
|
10
|
Martín-Rivada Á, Guerra-Cantera S, Campillo-Calatayud A, Andrés-Esteban EM, Sánchez Holgado M, Martos-Moreno GÁ, Pozo J, Güemes M, Soriano-Guillén L, Pellicer A, Oxvig C, Frystyk J, Chowen JA, Barrios V, Argente J. Pappalysins and Stanniocalcins and Their Relationship With the Peripheral IGF Axis in Newborns and During Development. J Clin Endocrinol Metab 2022; 107:2912-2924. [PMID: 35902207 DOI: 10.1210/clinem/dgac453] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Pappalysins (PAPP-A, PAPP-A2) modulate body growth by increasing insulin-like growth factor I (IGF-I) bioavailability through cleavage of insulin-like growth factor binding proteins (IGFBPs) and are inhibited by stanniocalcins (STC1, STC2). Normative data on these novel factors, as well as on free IGF-I and uncleaved fractions of IGFBPs, are not well established. OBJECTIVE This work aimed to determine serum concentrations of PAPP-A, PAPP-A2, STC1, and STC2 in relationship with other growth hormone (GH)-IGF axis parameters during development. METHODS Full-term newborns (150; gestational age: 39.30 ± 1.10 weeks), 40 preterm newborns (30.87 ± 3.35 weeks), and 1071 healthy individuals (aged 1-30 years) were included in the study and divided according to their Tanner stages (males and females): I:163 males, 154 females; II:100 males, 75 females; III:83 males, 96 females; IV: 77 males, 86 females; and V:109 males,128 females. RESULTS Serum concentrations of PAPP-A, PAPP-A2, STC1, STC2, IGFBP-2, total IGFBP-4, and total IGFBP-5 were elevated at birth and declined throughout childhood. In postnatal life, PAPP-A2 concentrations decreased progressively in concomitance with the free/total IGF-I ratio; however, stanniocalcin concentrations remained stable. PAPP-A2 concentrations positively correlated with the free/total IGF-I ratio (r = +0.28; P < .001) and negatively with the intact/total IGFBP-3 ratio (r = -0.23; P < .001). PAPP-A concentrations inversely correlated with intact/total IGFBP-4 ratio (r = -0.21; P < .001), with PAPP-A concentrations being lower in females at all ages. Association studies indicate the importance of stanniocalcins and pappalysins in the control of this axis in an age-specific manner. CONCLUSION This study provides reference values of pappalysins and stanniocalcins, which modulate IGF-I activity by changing the concentrations of cleaved and uncleaved IGFBPs.
Collapse
Affiliation(s)
- Álvaro Martín-Rivada
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
| | - Santiago Guerra-Cantera
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - Ana Campillo-Calatayud
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
| | | | | | - Gabriel Á Martos-Moreno
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - Jesús Pozo
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - María Güemes
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
| | - Leandro Soriano-Guillén
- Hospital Universitario Fundación Jiménez Díaz, Instituto de Investigación Fundación Jiménez Díaz, E-28040, Madrid, Spain
| | - Adelina Pellicer
- Department of Neonatology, Hospital Universitario La Paz, E-28046, Madrid, Spain
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, DK-8000, Aarhus C, Aarhus, Denmark
| | - Jan Frystyk
- Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Julie A Chowen
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, E-28049, Madrid, Spain
| | - Vicente Barrios
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
| | - Jesús Argente
- Hospital Infantil Universitario Niño Jesús, Departments of Pediatrics & Pediatric Endocrinology, Research Institute "La Princesa," Department of Pediatrics, Universidad Autónoma de Madrid, E-28009, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, E-28009, Madrid, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, E-28049, Madrid, Spain
| |
Collapse
|
11
|
Gómez-Cuadrado L, Bullock E, Mabruk Z, Zhao H, Souleimanova M, Noer PR, Turnbull AK, Oxvig C, Bertos N, Byron A, Dixon JM, Park M, Haider S, Natrajan R, Sims AH, Brunton VG. Characterisation of the Stromal Microenvironment in Lobular Breast Cancer. Cancers (Basel) 2022; 14:904. [PMID: 35205651 PMCID: PMC8870100 DOI: 10.3390/cancers14040904] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 02/06/2022] [Indexed: 11/16/2022] Open
Abstract
Invasive lobular carcinoma (ILC) is the second most common histological subtype of breast cancer, and it exhibits a number of clinico-pathological characteristics distinct from the more common invasive ductal carcinoma (IDC). We set out to identify alterations in the tumor microenvironment (TME) of ILC. We used laser-capture microdissection to separate tumor epithelium from stroma in 23 ER+ ILC primary tumors. Gene expression analysis identified 45 genes involved in regulation of the extracellular matrix (ECM) that were enriched in the non-immune stroma of ILC, but not in non-immune stroma from ER+ IDC or normal breast. Of these, 10 were expressed in cancer-associated fibroblasts (CAFs) and were increased in ILC compared to IDC in bulk gene expression datasets, with PAPPA and TIMP2 being associated with better survival in ILC but not IDC. PAPPA, a gene involved in IGF-1 signaling, was the most enriched in the stroma compared to the tumor epithelial compartment in ILC. Analysis of PAPPA- and IGF1-associated genes identified a paracrine signaling pathway, and active PAPP-A was shown to be secreted from primary CAFs. This is the first study to demonstrate molecular differences in the TME between ILC and IDC identifying differences in matrix organization and growth factor signaling pathways.
Collapse
Affiliation(s)
- Laura Gómez-Cuadrado
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Esme Bullock
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Zeanap Mabruk
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Hong Zhao
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Margarita Souleimanova
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark; (P.R.N.); (C.O.)
| | - Arran K. Turnbull
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, DK-8000 Aarhus C, Denmark; (P.R.N.); (C.O.)
| | - Nicholas Bertos
- Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada;
| | - Adam Byron
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - J. Michael Dixon
- Edinburgh Breast Unit, University of Edinburgh, Edinburgh EH4 2XU, UK;
| | - Morag Park
- Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada; (H.Z.); (M.S.); (M.P.)
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.H.); (R.N.)
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.H.); (R.N.)
| | - Andrew H. Sims
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| | - Valerie G. Brunton
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XR, UK; (L.G.-C.); (E.B.); (Z.M.); (A.K.T.); (A.B.)
| |
Collapse
|
12
|
Rojas-Rodriguez R, Ziegler R, DeSouza T, Majid S, Madore AS, Amir N, Pace VA, Nachreiner D, Alfego D, Mathew J, Leung K, Moore Simas TA, Corvera S. PAPPA-mediated adipose tissue remodeling mitigates insulin resistance and protects against gestational diabetes in mice and humans. Sci Transl Med 2020; 12:eaay4145. [PMID: 33239385 PMCID: PMC8375243 DOI: 10.1126/scitranslmed.aay4145] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 04/25/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022]
Abstract
Pregnancy is a physiological state of continuous adaptation to changing maternal and fetal nutritional needs, including a reduction of maternal insulin sensitivity allowing for appropriately enhanced glucose availability to the fetus. However, excessive insulin resistance in conjunction with insufficient insulin secretion results in gestational diabetes mellitus (GDM), greatly increasing the risk for pregnancy complications and predisposing both mothers and offspring to future metabolic disease. Here, we report a signaling pathway connecting pregnancy-associated plasma protein A (PAPPA) with adipose tissue expansion in pregnancy. Adipose tissue plays a central role in the regulation of insulin sensitivity, and we show that, in both mice and humans, pregnancy caused remodeling of adipose tissue evidenced by altered adipocyte size, vascularization, and in vitro expansion capacity. PAPPA is known to be a metalloprotease secreted by human placenta that modulates insulin-like growth factor (IGF) bioavailability through prolteolysis of IGF binding proteins (IGFBPs) 2, 4, and 5. We demonstrate that recombinant PAPPA can stimulate ex vivo human adipose tissue expansion in an IGFBP-5- and IGF-1-dependent manner. Moreover, mice lacking PAPPA displayed impaired adipose tissue remodeling, pregnancy-induced insulin resistance, and hepatic steatosis, recapitulating multiple aspects of human GDM. In a cohort of 6361 pregnant women, concentrations of circulating PAPPA are inversely correlated with glycemia and odds of developing GDM. These data identify PAPPA and the IGF signaling pathway as necessary for the regulation of maternal adipose tissue physiology and systemic glucose homeostasis, with consequences for long-term metabolic risk and potential for therapeutic use.
Collapse
Affiliation(s)
- Raziel Rojas-Rodriguez
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rachel Ziegler
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sana Majid
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Aylin S Madore
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Nili Amir
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Veronica A Pace
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Daniel Nachreiner
- Clinical Translational Research Pathway, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David Alfego
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Jomol Mathew
- Division of Data Sciences and Technology, IT, University of Massachusetts Medical School, Worcester, MA 01605, USA
- Department of Population and Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Katherine Leung
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Tiffany A Moore Simas
- Departments of Obstetrics and Gynecology, University of Massachusetts Medical School and UMass Memorial Healthcare, Worcester, MA 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
13
|
Jenkins EC, Brown SO, Germain D. The Multi-Faced Role of PAPP-A in Post-Partum Breast Cancer: IGF-Signaling is Only the Beginning. J Mammary Gland Biol Neoplasia 2020; 25:181-189. [PMID: 32901383 DOI: 10.1007/s10911-020-09456-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/24/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling and control of local bioavailability of free IGF by the IGF binding proteins (IGFBP) are important regulators of both mammary development and breast cancer. A recent genome-wide association study (GWAS) identified small nucleotide polymorphisms that reduce the expression of IGFBP-5 as a risk factor of developing breast cancer. This observation suggests that genetic alterations leading to a decreased level of IGFBP-5 may also contribute to breast cancer. In the current review, we focus on Pregnancy-Associated Plasma Protein A (PAPP-A), a protease involved in the degradation of IGFBP-5. PAPP-A is overexpressed in the majority of breast cancers but its role in cancer has only begun to be explored. More specifically, this review aims at highlighting the role of post-partum involution in the oncogenic function of PAPP-A. Notably, we summarize recent studies indicating that PAPP-A plays a role not only in the degradation of IGFBP-5 but also in the deposition of collagen and activation of the collagen receptor discoidin 2 (DDR2) during post-partum involution. Finally, considering the immunosuppressive microenvironment of post-partum involution, we also discuss the unexpected finding made in Ewing Sarcoma that PAPP-A plays a role in immune evasion. While the immunosuppressive role of PAPP-A in breast cancer remains to be determined, collectively these studies highlight the multifaced role of PAPP-A in cancer that extends well beyond its effect on IGF-signaling.
Collapse
Affiliation(s)
- Edmund Charles Jenkins
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Samantha O Brown
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA
| | - Doris Germain
- Department of Medicine, Division of Hematology/ Oncology, Icahn School of Medicine at Mount Sinai, Tisch Cancer Institute, New York, NY, 10029, USA.
| |
Collapse
|
14
|
Frystyk J, Teran E, Gude MF, Bjerre M, Hjortebjerg R. Pregnancy-associated plasma proteins and Stanniocalcin-2 - Novel players controlling IGF-I physiology. Growth Horm IGF Res 2020; 53-54:101330. [PMID: 32693362 DOI: 10.1016/j.ghir.2020.101330] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 10/23/2022]
Abstract
IGF-I was originally discovered as a GH-dependent growth factor stimulating longitudinal growth. Currently, however, it has become evident that the biological activities of IGF-I extend well beyond those of a simple growth factor and impact such processes as insulin sensitivity, aging, cancer and cardiovascular disease. The vast majority of IGF-I is tightly bound to IGF-binding proteins (IGFBPs), which renders IGF-I unable to stimulate the IGF-I receptor (IGF-IR) in vivo. This binding means that liberation of IGF-I from the IGFBPs is an important step controlling IGF-I action. In this context, IGFBP-cleaving enzymes appear to play a key role. Enzymatic cleavage of the IGFBPs markedly lowers their ligand affinity, and as a consequence, IGF-I becomes liberated and hence available for stimulation of the IGF-IR. Two of the best-characterized IGFBP-cleaving enzymes are pregnancy-associated plasma protein-A (PAPP-A) and its paralog PAPP-A2. The two enzymes (often referred to as pappalysins) regulate the liberation of IGF-I in a highly controlled manner. PAPP-A is believed to act predominantly in tissues, serving to liberate IGF-I at the cell surface in close proximity to the IGF-IR. In keeping with this notion, mice lacking PAPP-A exhibit reduced body size, despite having normal circulating IGF-I concentrations. In contrast, human findings indicate that altered PAPP-A2 activity changes circulating IGF-I concentrations, although PAPP-A2 is also present in high concentrations in tissues. Thus, PAPP-A2 appears to impact circulating, as well as tissue, IGF-I activity. The enzymatic activity of PAPP-A and PAPP-A2 was recently discovered to be regulated by the protein Stanniocalcin-2 (STC2). By binding to the enzymatic sites of PAPP-A and PAPP-A2, STC2 inhibits their activity. To date, the majority of findings demonstrating the ability of pappalysins and STC2 to regulate IGF-I action are from preclinical studies. However, clinical studies are now beginning to emerge. In this review, we will summarize our data on STC2, PAPP-A and PAPP-A2 in humans. These results indicate that pappalysins and STC2 constitute an important IGF-I activity-regulating system that warrants further investigation.
Collapse
Affiliation(s)
- Jan Frystyk
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.
| | - Enrique Teran
- Colegio de Ciencias de la Salud, Universidad San Francisco de Quito, Quito, Ecuador
| | - Mette Faurholdt Gude
- Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Health, Aarhus University, Aarhus, Denmark
| | - Rikke Hjortebjerg
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital & Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Steno Diabetes Center Odense (SDCO), Odense University Hospital, Odense, Denmark
| |
Collapse
|
15
|
Adams AD, Guedj F, Bianchi DW. Placental development and function in trisomy 21 and mouse models of Down syndrome: Clues for studying mechanisms underlying atypical development. Placenta 2020; 89:58-66. [PMID: 31683073 PMCID: PMC10040210 DOI: 10.1016/j.placenta.2019.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 12/17/2022]
Abstract
Down syndrome (DS) is the most common genetic disorder leading to developmental disability. The phenotypes associated with DS are complex and vary between affected individuals. Placental abnormalities in DS include differences in cytotrophoblast fusion that affect subsequent conversion to syncytiotrophoblast, atypical oxidative stress/antioxidant balance, and increased expression of genes that are also upregulated in the brains of individuals with Alzheimer's disease. Placentas in DS are prematurely senescent, showing atypical evidence of mineralization. Fetuses with DS are especially susceptible to adverse obstetric outcomes, including early in utero demise, stillbirth and growth restriction, all of which are related to placental function. The placenta, therefore, may provide key insights towards understanding the phenotypic variability observed in individuals with DS and aid in identifying biomarkers that can be used to evaluate phenotypic severity and prenatal treatments in real time. To address these issues, many different mouse models of DS have been generated to identify the mechanisms underlying developmental changes in many organ systems. Little is known, however, regarding placental development in the currently available mouse models of DS. Based upon the relative paucity of data on placental development in preclinical mouse models of DS, we recommend that future evaluation of new and existing models routinely include histologic and functional assessments of the placenta. In this paper we summarize studies performed in the placentas of both humans and mouse models with DS, highlighting gaps in knowledge and suggesting directions for future research.
Collapse
Affiliation(s)
- April D Adams
- Medical Genetics Branch (Prenatal Genomics and Therapy Section), National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Faycal Guedj
- Medical Genetics Branch (Prenatal Genomics and Therapy Section), National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Diana W Bianchi
- Medical Genetics Branch (Prenatal Genomics and Therapy Section), National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA; Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Tuunainen E, Lund J, Danielsson J, Pietilä P, Wahlroos V, Pudge K, Leinonen I, Porela P, Ilva T, Lepäntalo M, Pulkki K, Voipio-Pulkki LM, Pettersson K, Wittfooth S. Direct Immunoassay for Free Pregnancy-Associated Plasma Protein A (PAPP-A). J Appl Lab Med 2019; 3:438-449. [DOI: 10.1373/jalm.2018.026096] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/27/2018] [Indexed: 11/06/2022]
Abstract
Abstract
Background
Pregnancy-associated plasma protein A (PAPP-A), especially in its noncomplexed form (fPAPP-A), is linked to vulnerable atherosclerotic plaques and risk of cardiac events. An assay for sensitive detection of fPAPP-A has been lacking. Our aim was to develop and validate a direct fPAPP-A assay to meet this need.
Methods
Monoclonal antibodies binding exclusively fPAPP-A were produced by immunizing mice with recombinant PAPP-A. In the optimized immunoassay, we used an fPAPP-A–specific capture antibody together with a lanthanide-chelate–labeled monoclonal antibody recognizing all PAPP-A forms. The assay was evaluated with CLSI guidelines and compared to a 2-assay subtractive fPAPP-A approach. Clinical performance was assessed with acute coronary syndrome patients.
Results
The limits of detection and quantitation were 0.4 mIU/L and 1.3 mIU/L, respectively, and the assay was linear up to 1000 mIU/L (R2 = 0.999). Both serum and heparin plasma were suitable matrices, and the complexed form of PAPP-A caused no significant interference. Correlation between the developed assay and the 2-assay approach was fair (Pearson's r = 0.819). Median concentration in healthy individuals was 1.0 mIU/L. fPAPP-A concentration was higher in patients who had myocardial infarction or died during the 1-year follow-up period than in those who did not (1.13 mIU/L vs 0.82 mIU/L, P = 0.008, model adjusted with age and sex). fPAPP-A measured with this direct assay predicted this end point as well as (follow-up 1 year) or better (30 days) than the 2-assay fPAPP-A alone or in combination with cTnI.
Conclusions
The new assay enables sensitive and reliable measurement of low cardiac-related fPAPP-A concentrations from blood samples.
Collapse
Affiliation(s)
- Emilia Tuunainen
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Juha Lund
- Heart Center, Turku University Hospital, Turku, Finland
| | - Joanna Danielsson
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Pirjo Pietilä
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Veikko Wahlroos
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Keira Pudge
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Isto Leinonen
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Pekka Porela
- Heart Center, Turku University Hospital, Turku, Finland
| | - Tuomo Ilva
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere, Finland
| | - Mauri Lepäntalo
- Department of Vascular Surgery, Helsinki University Central Hospital, Helsinki, Finland
| | - Kari Pulkki
- Laboratory Division, Turku University Hospital, Turku, Finland
| | | | - Kim Pettersson
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| | - Saara Wittfooth
- Department of Biochemistry/Biotechnology, University of Turku, Turku, Finland
| |
Collapse
|
17
|
Dobolyi A, Lékó AH. The insulin-like growth factor-1 system in the adult mammalian brain and its implications in central maternal adaptation. Front Neuroendocrinol 2019; 52:181-194. [PMID: 30552909 DOI: 10.1016/j.yfrne.2018.12.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 11/04/2018] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Our knowledge on the bioavailability and actions of insulin-like growth factor-1 (IGF-1) has markedly expanded in recent years as novel mechanisms were discovered on IGF binding proteins (IGFBPs) and their ability to release IGF-1. The new discoveries allowed a better understanding of the endogenous physiological actions of IGF-1 and also its applicability in therapeutics. The focus of the present review is to summarize novel findings on the neuronal, neuroendocrine and neuroplastic actions of IGF-1 in the adult brain. As most of the new regulatory mechanisms were described in the periphery, their implications on brain IGF system will also be covered. In addition, novel findings on the effects of IGF-1 on lactation and maternal behavior are described. Based on the enormous neuroplastic changes related to the peripartum period, IGF-1 has great but largely unexplored potential in maternal adaptation of the brain, which is highlighted in the present review.
Collapse
Affiliation(s)
- Arpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary.
| | - András H Lékó
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Hungarian Academy of Sciences and Eötvös Loránd University, Budapest, Hungary; Laboratory of Neuromorphology, Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary; Department of Psychiatry and Psychotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
18
|
Hjortebjerg R. IGFBP-4 and PAPP-A in normal physiology and disease. Growth Horm IGF Res 2018; 41:7-22. [PMID: 29864720 DOI: 10.1016/j.ghir.2018.05.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/15/2018] [Accepted: 05/29/2018] [Indexed: 02/07/2023]
Abstract
Insulin-like growth factor (IGF) binding protein-4 (IGFBP-4) is a modulator of the IGF system, exerting both inhibitory and stimulatory effects on IGF-induced cellular growth. IGFBP-4 is the principal substrate for the enzyme pregnancy-associated plasma protein-A (PAPP-A). Through IGF-dependent cleavage of IGFBP-4 in the vicinity of the IGF receptor, PAPP-A is able to increase IGF bioavailability and stimulate IGF-mediated growth. Recently, the stanniocalcins (STCs) were identified as novel inhibitors of PAPP-A proteolytic activity, hereby adding additional members to the seemingly endless list of proteins belonging to the IGF family. Our understanding of these proteins has advanced throughout recent years, and there is evidence to suggest that the role of IGFBP-4 and PAPP-A in defining the relationship between total IGF and IGF bioactivity can be linked to a number of pathological conditions. This review provides an overview of the experimental and clinical findings on the IGFBP-4/PAPP-A/STC axis as a regulator of IGF activity and examines the conundrum surrounding extrapolation of circulating concentrations to tissue action of these proteins. The primary focus will be on the biological significance of IGFBP-4 and PAPP-A in normal physiology and in pathophysiology with emphasis on metabolic disorders, cardiovascular diseases, and cancer. Finally, the review assesses current new trajectories of IGFBP-4 and PAPP-A research.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| |
Collapse
|
19
|
Petry CJ, Ong KK, Hughes IA, Acerini CL, Dunger DB. The association between age at menarche and later risk of gestational diabetes is mediated by insulin resistance. Acta Diabetol 2018; 55:853-859. [PMID: 29789944 PMCID: PMC6060956 DOI: 10.1007/s00592-018-1162-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/15/2018] [Indexed: 02/07/2023]
Abstract
AIMS Associations have been reported between age at menarche and the later risk of gestational diabetes. However, it is not known whether these associations reflect differences in insulin sensitivity and/or pancreatic β-cell function in pregnancy. METHODS We examined this question in women enrolled in the prospective Cambridge Baby Growth Study who recalled their age at menarche in questionnaires during pregnancy. Polynomial logistic and linear regression models were used to relate menarche timing to the risk of gestational diabetes, both unadjusted and adjusted for the Homeostasis Model Assessments of insulin resistance (HOMA IR) and pancreatic β-cell function (HOMA B) at week 28 of pregnancy. RESULTS Age at menarche showed a U-shaped association with gestational diabetes risk (linear term: p = 9.5 × 10-4; quadratic term: p = 1.0 × 10-3; n = 889; overall model p = 8.1 × 10-3). Age at menarche showed a negative linear association with insulin resistance (HOMA IR: β = -0.13, p = 5.2 × 10-4, n = 771), which explained the relationship between age at menarche and gestational diabetes risk (adjusted linear term going from p = 0.03-0.08; adjusted quadratic term going from p = 0.04-0.08; n = 771). Age at menarche also showed a negative linear association with β-cell function (HOMA B: β = -0.11, p = 2.8 × 10-3, n = 771) but this did not attenuate the relationship between age at menarche and gestational diabetes (adjusted linear term p = 0.02; adjusted quadratic term p = 0.03, n = 771). CONCLUSIONS These results suggest that the associations between age at menarche and risk of gestational diabetes and raised pregnancy glucose concentrations may be mediated by insulin resistance.
Collapse
Affiliation(s)
- Clive J Petry
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Box 116, Cambridge, CB2 0QQ, UK.
| | - Ken K Ong
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Box 116, Cambridge, CB2 0QQ, UK
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK
- The Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Ieuan A Hughes
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Box 116, Cambridge, CB2 0QQ, UK
| | - Carlo L Acerini
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Box 116, Cambridge, CB2 0QQ, UK
| | - David B Dunger
- Department of Paediatrics, University of Cambridge, Cambridge Biomedical Campus, Hills Road, Box 116, Cambridge, CB2 0QQ, UK
- The Institute of Metabolic Science, University of Cambridge, Cambridge, CB2 0QQ, UK
| |
Collapse
|
20
|
Abstract
The zinc metalloproteinase, PAPP-A, enhances local insulin-like growth factor (IGF) action through cleavage of inhibitory IGF-binding proteins, thereby increasing IGF available for IGF receptor-mediated cell proliferation, migration and survival. In many tumors, enhanced IGF receptor signaling is associated with tumor growth, invasion and metastasis. We will first discuss PAPP-A structure and function, and post-translational inhibitors of PAPP-A expression or proteolytic activity. We will then review the evidence supporting an important role for PAPP-A in many cancers, including breast, ovarian and lung cancer, and Ewing sarcoma.
Collapse
Affiliation(s)
- Cheryl A Conover
- From the Division of Endocrinology Mayo ClinicRochester, Minnesota, USA
| | - Claus Oxvig
- Department of Molecular Biology and GeneticsAarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Barile L, Cervio E, Lionetti V, Milano G, Ciullo A, Biemmi V, Bolis S, Altomare C, Matteucci M, Di Silvestre D, Brambilla F, Fertig TE, Torre T, Demertzis S, Mauri P, Moccetti T, Vassalli G. Cardioprotection by cardiac progenitor cell-secreted exosomes: role of pregnancy-associated plasma protein-A. Cardiovasc Res 2018. [DOI: 10.1093/cvr/cvy055] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Lucio Barile
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Elisabetta Cervio
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna and UOS Anesthesiology, Fondazione Toscana G. Monasterio, Pisa, Italy
| | - Giuseppina Milano
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
- Heart and Vessels Department, CHUV-University of Lausanne Medical Hospital, Lausanne, Switzerland
| | - Alessandra Ciullo
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Vanessa Biemmi
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Sara Bolis
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Claudia Altomare
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Marco Matteucci
- Institute of Life Sciences, Scuola Superiore Sant'Anna and UOS Anesthesiology, Fondazione Toscana G. Monasterio, Pisa, Italy
| | | | | | | | - Tiziano Torre
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Stefanos Demertzis
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | | | - Tiziano Moccetti
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
| | - Giuseppe Vassalli
- Cellular and Molecular Cardiology Laboratory, Cardiocentro Ticino Foundation and Swiss Institute for Regenerative Medicine (SIRM), via Tesserete 48, 6900 Lugano, Switzerland
- Heart and Vessels Department, CHUV-University of Lausanne Medical Hospital, Lausanne, Switzerland
- Department of Cardiology, Molecular Cardiology Institute, University of Zürich, Zürich, Switzerland
| |
Collapse
|
22
|
Espelund US, Bjerre M, Hjortebjerg R, Rasmussen TR, Lundby A, Hoeflich A, Folkersen BH, Oxvig C, Frystyk J. Insulin-Like Growth Factor Bioactivity, Stanniocalcin-2, Pregnancy-Associated Plasma Protein-A, and IGF-Binding Protein-4 in Pleural Fluid and Serum From Patients With Pulmonary Disease. J Clin Endocrinol Metab 2017; 102:3526-3534. [PMID: 28911149 DOI: 10.1210/jc.2017-00033] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 07/11/2017] [Indexed: 01/02/2023]
Abstract
CONTEXT Members of the insulin-like growth factor (IGF) system are primarily produced in the liver and secreted into the circulation, but they are also produced, recruited, and activated locally in tissues. OBJECTIVE To compare activity and concentrations of IGF system components in pleural fluid and blood. DESIGN Pathological pleural fluid, secondary to lung cancer or nonmalignant disease, and matching blood samples were collected from 24 patients ages 66.7 to 81.9 years. METHODS IGF-related proteins and cytokine levels were measured by immunoassays or immunoblotting. Bioactive IGF was measured by an IGF-1 receptor phosphorylation assay. RESULTS Total IGF-1 concentration did not differ between the compartments, but concentrations of free IGF-1 and bioactive IGF were more than threefold higher in pleural fluid than in corresponding serum samples (P = 0.0004), regardless of etiology. Median pregnancy-associated plasma protein-A (PAPP-A) and interleukin (IL)-6 levels were increased 47-fold and 143-fold, respectively, in pleural fluid compared with plasma (P < 0.0001). PAPP-A and IL-6 concentrations correlated positively (r = 0.46; P = 0.02). In pleural fluid, levels of PAPP-A-generated IGF binding protein-4 fragments correlated inversely with that of stanniocalcin-2 (r ≤ -0.42; P ≤ 0.05), a PAPP-A inhibitor; such correlations were absent in plasma. CONCLUSION Pathological pleural fluid is characterized by increased in vitro IGF bioactivity and elevated concentrations of PAPP-A, an IGF-activating proteinase. Thus, the tissue activity of the IGF system may differ substantially from that of the circulating IGF system. The correlation between IL-6 and PAPP-A indicates that inflammation plays a role in promoting local tissue IGF activity.
Collapse
Affiliation(s)
- Ulrick Skipper Espelund
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
- Danish Diabetes Academy, DK-5000 Odense C, Denmark
| | - Torben Riis Rasmussen
- Department of Pulmonary Medicine, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| | - Anders Lundby
- Safety and GI Biology, Novo Nordisk A/S, Novo Nordisk Park, G9.S.17, DK-2760 Måløv, Denmark
| | - Andreas Hoeflich
- Institute of Genome Biology, Leibniz-Institute for Farm Animal Biology, D-18196 Dummerstorf, Germany
| | | | - Claus Oxvig
- Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, DK-8000 Aarhus C, Denmark
| |
Collapse
|
23
|
Petry CJ, Ong KK, Hughes IA, Acerini CL, Frystyk J, Dunger DB. Early Pregnancy-Associated Plasma Protein A Concentrations Are Associated With Third Trimester Insulin Sensitivity. J Clin Endocrinol Metab 2017; 102:2000-2008. [PMID: 28323969 PMCID: PMC5464396 DOI: 10.1210/jc.2017-00272] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 03/08/2017] [Indexed: 01/04/2023]
Abstract
CONTEXT First or early second trimester pregnancy-associated plasma protein A (PAPP-A) concentrations have previously been shown to be lower in women who subsequently develop gestational diabetes mellitus (GDM) and gestational hypertension. OBJECTIVE We therefore sought to investigate why circulating PAPP-A concentrations are related to the subsequent risk of GDM and gestational hypertension. PATIENTS, DESIGN, AND SETTING We measured serum PAPP-A concentrations around week 15 of pregnancy and related these to indices derived from week 28 oral glucose tolerance tests and blood pressures across pregnancy in the Cambridge Baby Growth Study cohort. RESULTS Increased PAPP-A concentrations were associated with reduced GDM risk [odds ratio 0.623 (0.453, 0.856), P = 3.5 × 10-3, n = 777] and reduced mean arterial blood pressures (β = -0.202 to -0.177, P = 1.7 to 6.9 × 10-3, n = 347 to 355). They were also negatively associated with week 28 fasting (β = -0.149, P = 6.6 × 10-4, n = 777) and 60-minute (β = -0.188, P = 1.5 × 10-5, n = 777) oral glucose tolerance test glucose concentrations. These associations were underpinned by the strong associations between increased week 15 PAPP-A concentrations and decreased week 28 insulin resistance (homeostasis model assessment of insulin resistance: β = -0.319, P = 1.7 × 10-13, n = 768), as well as increased insulin secretion relative to insulin sensitivity (insulin disposition index: β = 0.202, P = 6.5 × 10-6, n = 731). CONCLUSIONS These results suggest that links between PAPP-A concentrations in early pregnancy and subsequent glucose concentrations and blood pressures may be mediated by changes in insulin sensitivity (and secretion).
Collapse
Affiliation(s)
- Clive J. Petry
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Ken K. Ong
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Medical Research Council Epidemiology Unit, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Ieuan A. Hughes
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Carlo L. Acerini
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Aarhus University, 8000 Aarhus, Denmark
| | - David B. Dunger
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
- Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
24
|
Kløverpris S, Mikkelsen JH, Pedersen JH, Jepsen MR, Laursen LS, Petersen SV, Oxvig C. Stanniocalcin-1 Potently Inhibits the Proteolytic Activity of the Metalloproteinase Pregnancy-associated Plasma Protein-A. J Biol Chem 2015. [PMID: 26195635 DOI: 10.1074/jbc.m115.650143] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
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.
Collapse
Affiliation(s)
| | | | | | | | | | - Steen V Petersen
- the Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark
| | - Claus Oxvig
- From the Department of Molecular Biology and Genetics and
| |
Collapse
|
25
|
The role of PAPP-A in the IGF system: location, location, location. J Cell Commun Signal 2015; 9:177-87. [PMID: 25617049 DOI: 10.1007/s12079-015-0259-9] [Citation(s) in RCA: 138] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
Although discovered as a placental protein present abundantly in the circulation of pregnant women, pregnancy-associated plasma protein-A (PAPP-A) is widely expressed in multiple tissues. PAPP-A is a highly specific metalloproteinase binding tightly to glycosaminoglycans present on the surface of cells. By cleaving a subset of insulin-like growth factor binding proteins (IGFBPs), PAPP-A thus functions within tissues as a growth-promoting enzyme, releasing bioactive IGF in close proximity to the IGF receptor. IGFBP-4 is believed to be the principal PAPP-A substrate, and the focus in this review is on PAPP-A enzymatic activity and its role in the PAPP-A-IGFBP-4-IGF axis, which is subject to regulation at several different levels. These include e.g., transcriptional control, competing reactions potentially sequestering IGF from IGFBP-4 and hence antagonizing PAPP-A-mediated IGF activation, and proteolytic inhibition of PAPP-A. The latter may involve the protein stanniocalcin-2 (STC2), recently found to potently inhibit PAPP-A activity by forming a covalent complex with PAPP-A. PAPP-A or complex-bound variants may escape from pathological tissues into the circulation. It is emphasized that the potential use of PAPP-A as a diagnostic or predictive biomarker in nonpregnant individuals requires precise knowledge of analyte identity and assay specificity in addition to an appropriate material for standardization. Finally, PAPP-A may serve as a therapeutic target to indirectly inhibit IGF signaling in tissues where this is driven by increased PAPP-A activity. By taking advantage of the intricate interaction between PAPP-A and IGFBP-4, highly specific and selective inhibition of PAPP-A is possible.
Collapse
|
26
|
Hjortebjerg R, Lindberg S, Hoffmann S, Jensen JS, Oxvig C, Bjerre M, Frystyk J. PAPP-A and IGFBP-4 fragment levels in patients with ST-elevation myocardial infarction treated with heparin and PCI. Clin Biochem 2014; 48:322-8. [PMID: 25489725 DOI: 10.1016/j.clinbiochem.2014.11.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/04/2014] [Accepted: 11/27/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVES Circulating levels of pregnancy-associated plasma protein-A (PAPP-A) predict outcome in patients with acute coronary syndrome (ACS). Unfortunately, administration of heparin to patients with ACS increases circulating PAPP-A, probably by a detachment of PAPP-A from cell surfaces, inducing a considerable bias when using PAPP-A as a biomarker. It remains unknown whether PAPP-A-derived N- and C-terminal fragments of insulin-like growth factor binding protein-4 (NT-IGFBP-4/CT-IGFBP-4) are acutely affected by the increase in PAPP-A. METHODS We prospectively included 78 patients with ST-segment elevation myocardial infarction (STEMI) treated with percutaneous coronary intervention (PCI). Prior to PCI, patients were injected with 10,000IU of unfractionated heparin (UFH). Blood samples were collected immediately before PCI, but after UFH-injection, immediately after PCI and on day 1 and day 2. Plasma IGFBP-4, CT-IGFBP-4 and NT-IGFBP-4 levels were determined by specific, novel immunoassays, and PAPP-A and IGF-I by commercial immunoassays. RESULTS Plasma PAPP-A was strongly elevated upon STEMI, UFH-administration and PCI with mean concentrations (95%-confidence interval) pre-PCI, post-PCI, day 1, and day 2 of 13.0 (11.2;15.2), 14.8 (13.1;16.8), 1.03 (0.90;1.18), and 1.08 (0.92;1.28) μg/L, respectively (p<0.0001). Pre-PCI concentrations of IGFBP-4, CT-IGFBP-4 and NT-IGFBP-4 were 154 (142;166), 53 (47;60) and 136 (122;150) μg/L, and levels were unaltered post-PCI. Concentrations increased on day 1 by 63 (43;87)%, 69 (36;110)%, and 47 (21;79)%, respectively (p<0.0001), i.e. at a time point when PAPP-A levels had normalized. CONCLUSION Plasma IGFBP-4-fragment levels are not acutely altered in patients with STEMI treated with UFH and PCI. Thus, they possess potentials as prognostic markers in ACS patients.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; The Danish Diabetes Academy, Odense, Denmark.
| | - Søren Lindberg
- Department of Cardiology P, Gentofte University Hospital, Copenhagen, Denmark
| | - Søren Hoffmann
- Department of Cardiology P, Gentofte University Hospital, Copenhagen, Denmark
| | - Jan S Jensen
- Department of Cardiology P, Gentofte University Hospital, Copenhagen, Denmark; Institute of Clinical Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, Aarhus, Denmark
| | - Mette Bjerre
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark
| | - Jan Frystyk
- Medical Research Laboratory, Department of Clinical Medicine, Faculty of Health, Aarhus University, Aarhus, Denmark; Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
27
|
Mikkelsen JH, Steffensen LB, Oxvig C. Development of a recombinant antibody towards PAPP-A for immunohistochemical use in multiple animal species. J Immunol Methods 2013; 404:33-40. [PMID: 24333852 DOI: 10.1016/j.jim.2013.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 12/04/2013] [Accepted: 12/05/2013] [Indexed: 10/25/2022]
Abstract
The metalloproteinase, pregnancy-associated plasma protein-A (PAPP-A), is increasingly recognized as a modulator of insulin-like growth factor (IGF) signaling; it cleaves IGF binding proteins causing the release of bioactive IGF. Accumulating evidence supports an important role of PAPP-A in both normal physiology and under different pathological conditions. However, antibodies for the detection of PAPP-A in non-human tissues have been lacking, although needed for use with several animal models which are currently being developed. To develop a monoclonal antibody suitable for the immunohistochemical detection of PAPP-A, we therefore selected a phage-derived scFv antibody, PAC1, specifically recognizing an epitope of PAPP-A, which is highly conserved between multiple animal species. We first converted this antibody into bivalent IgG, and verified its ability to recognize PAPP-A in sections of formalin-fixed and paraffin-embedded tissue. For increased sensitivity, affinity maturation to sub-nanomolar affinity was then carried out. The resulting recombinant antibody, PAC1-D8-mIgG2a, detects PAPP-A specifically and sensitively in human tissue. In addition, this antibody allows detection of PAPP-A in non-human species. We demonstrate its usefulness for the visualization of PAPP-A in murine and porcine tissues.
Collapse
Affiliation(s)
- Jakob H Mikkelsen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Lasse B Steffensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, DK-8000 Aarhus C, Denmark.
| |
Collapse
|
28
|
Kløverpris S, Gaidamauskas E, Rasmussen LCV, Overgaard MT, Kronborg C, Knudsen UB, Christiansen M, Kumar A, Oxvig C. A robust immunoassay for pregnancy-associated plasma protein-A2 based on analysis of circulating antigen: establishment of normal ranges in pregnancy. Mol Hum Reprod 2013; 19:756-63. [PMID: 23804707 DOI: 10.1093/molehr/gat047] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pregnancy-associated plasma protein-A (PAPP-A) and PAPP-A2, two homologous metzincin metalloproteases, are both tightly linked to regulation within the insulin-like growth factor (IGF) system because of their specific cleavage of IGF binding proteins. Recent studies suggest that PAPP-A may be involved in clinical conditions related to unwanted cellular growth, and the circulating levels of PAPP-A is an established biomarker in prenatal screening for chromosomal abnormalities. Microarray data indicate that PAPP-A2 has potential as a biomarker for pre-eclampsia. However, well-characterized immunological methods of quantification are not available. We therefore developed monoclonal antibodies against recombinant PAPP-A2. The antibodies were epitope mapped against recombinantly expressed chimeras between PAPP-A2 and PAPP-A. Furthermore, circulating PAPP-A2 was immunoaffinity purified and characterized by sequence analysis and mass spectrometry. Unlike PAPP-A, PAPP-A2 is a noncovalent dimer in which each subunit of 1558 amino acids originates from all of the 22 predicted coding exons. A previously hypothesized variant (PAPP-E) does not exist, but low amounts of a C-terminally truncated PAPP-A2 variant was detected. A sensitive and robust ELISA for full-length PAPP-A2 was developed and used to establish normal ranges of PAPP-A2 through pregnancy. The functional sensitivity of this ELISA at 20% CV was 0.08 ng/ml, and the serum concentration of PAPP-A2 was found to increase during pregnancy in agreement with placental synthesis. The existence of this assay will enable an assessment of the biomarker potential of PAPP-A2 in pre-eclampsia as well as other clinical conditions.
Collapse
Affiliation(s)
- S Kløverpris
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, Aarhus C DK-8000, Denmark
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Formation of high-molecular-weight angiotensinogen during pregnancy is a result of competing redox reactions with the proform of eosinophil major basic protein. Biochem J 2013; 449:209-17. [PMID: 23033876 DOI: 10.1042/bj20120801] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The plasma concentration of the placentally derived proMBP (proform of eosinophil major basic protein) increases in pregnancy, and three different complexes containing proMBP have been isolated from pregnancy plasma and serum: a 2:2 complex with the metalloproteinase, PAPP-A (pregnancy-associated plasma protein-A), a 2:2 complex with AGT (angiotensinogen) and a 2:2:2 complex with AGT and complement C3dg. In the present study we show that during human pregnancy, all of the circulating proMBP exists in covalent complexes, bound to either PAPP-A or AGT. We also show that the proMBP-AGT complex constitutes the major fraction of circulating HMW (high-molecular weight) AGT in late pregnancy, and that this complex is able to further associate with complement C3 derivatives post-sampling. Clearance experiments in mice suggest that complement C3-based complexes are removed faster from the circulation compared to monomeric AGT and the proMBP-AGT complex. Furthermore, we have used recombinant proteins to analyse the formation of the proMBP-PAPP-A and the proMBP-AGT complexes, and we demonstrate that they are competing reactions, depending on the same cysteine residue of proMBP, but differentially on the redox potential, potentially important for the relative amounts of the complexes in vivo. These findings may be important physiologically, since the biochemical properties of the proteins change as a consequence of complex formation.
Collapse
|
30
|
Mikkelsen JH, Gyrup C, Kristensen P, Overgaard MT, Poulsen CB, Laursen LS, Oxvig C. Inhibition of the proteolytic activity of pregnancy-associated plasma protein-A by targeting substrate exosite binding. J Biol Chem 2008; 283:16772-80. [PMID: 18434323 DOI: 10.1074/jbc.m802429200] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The metalloproteinase pregnancy-associated plasma protein-A (PAPP-A) cleaves both insulin-like growth factor (IGF)-binding protein 4 (IGFBP-4) and -5 at a single site in their central domain causing the release of bioactive IGF. Inhibition of IGF signaling is relevant in human disease, and several drugs in development target the IGF receptor. However, inhibition of PAPP-A activity may be a valuable alternative. We have generated monoclonal phage-derived single chain fragment variable (scFv) antibodies which selectively inhibit the cleavage of IGFBP-4 by PAPP-A, relevant under conditions where cleavage of IGFBP-4 represents the final step in the delivery of IGF to the IGF receptor. None of the antibodies inhibited the homologous proteinase PAPP-A2, which allowed mapping of antibody binding by means of chimeras between PAPP-A and PAPP-A2 to the C-terminal Lin12-Notch repeat module, separated from the proteolytic domain by almost 1000 amino acids. Hence, the antibodies define a substrate binding exosite that can be targeted for the selective inhibition of PAPP-A proteolytic activity against IGFBP-4. In addition, we show that the Lin12-Notch repeat module reversibly binds a calcium ion and that bound calcium is required for antibody binding, providing a strategy for the further development of selective inhibitory compounds. To our knowledge these data represent the first example of differential inhibition of cleavage of natural proteinase substrates by exosite targeting. Generally, exosite inhibitors are less likely to affect the activity of related proteolytic enzymes with similar active site environments. In the case of PAPP-A, selective inhibition of IGFBP-4 cleavage by interference with exosite binding is a further advantage, as the activity against other known or unknown PAPP-A substrates, whose cleavage may not depend on binding to the same exosite, is not targeted.
Collapse
Affiliation(s)
- Jakob H Mikkelsen
- Department of Molecular Biology, University of Aarhus, DK-8000 Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|