1
|
Bøtkjær JA, Poulsen LLC, Noer PR, Grøndahl ML, Englund ALM, Franks S, Hardy K, Oxvig C, Andersen CY. Dynamics of IGF Signaling During the Ovulatory Peak in Women Undergoing Ovarian Stimulation. J Clin Endocrinol Metab 2024; 110:e160-e167. [PMID: 38436415 DOI: 10.1210/clinem/dgae132] [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: 11/13/2023] [Revised: 02/04/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024]
Abstract
CONTEXT Insulin-like growth factor (IGF) signaling is known to affect human ovarian follicular function during growth and development. However, the role of the IGF system is unknown during the ovulatory peak, which is characterized by profound changes in granulosa cell (GCs) mitosis and function. OBJECTIVE How is the IGF system expressed and regulated during the midcycle surge in women? METHODS Follicular fluid (FF) and GCs were collected during the ovulatory peak from 2 specific time points. One sample was obtained before oocyte pickup (OPU): before ovulation trigger (OT) (T = 0 hours) or at 12, 17, or 32 hours after OT, and 1 sample was obtained at OPU 36 hours after OT. Fifty women undergoing ovarian stimulation at a university hospital were included. Gene expression profiles were assessed by microarray analysis of GCs. IGF-related proteins in the FF were assessed by immunoassay or by determination of activity with a proteinase assay. RESULTS Gene expression of proteins promoting IGF activity (ie, IGF2, PAPP-A, and IRS1) together with proliferation markers were downregulated on a transcriptional level in GCs after OT, whereas proteins inhibiting the IGF signal (ie, IGFBPs, IGF2, and STC1) were upregulated. STC1 gene expression and protein levels were greatly upregulated after OT with a parallel steep downregulation of PAPP-A proteolytic activity. CONCLUSION These data suggest that downregulation of IGF signaling mediated by increased STC1 expression is instrumental for the sudden cessation in GC proliferation and onset of differentiation during the ovulatory peak.
Collapse
Affiliation(s)
- Jane Alrø Bøtkjær
- Laboratory of Reproductive Biology, University Hospital of Copenhagen, Rigshospitalet, 2100 Copenhagen, Denmark
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Liv la Cour Poulsen
- Fertility Clinic, Zealand University Hospital, 4600 Køge, Denmark
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Pernille Rimmer Noer
- Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus, Denmark
| | - Marie Louise Grøndahl
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | | | - Stephen Franks
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Kate Hardy
- Institute of Reproductive and Developmental Biology, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, University of Aarhus, 8000 Aarhus, Denmark
| | - Claus Yding Andersen
- Fertility Clinic, University Hospital of Copenhagen, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
2
|
Jia K, Cao L, Yu Y, Jing D, Wu W, Van Tine BA, Shao Z. Signaling pathways and targeted therapies in Ewing sarcoma. Pharmacol Ther 2024; 266:108765. [PMID: 39622389 DOI: 10.1016/j.pharmthera.2024.108765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Ewing sarcoma, the second most prevalent malignant bone tumor with potential occurrence in soft tissues, exhibits a high level of aggressiveness, primarily afflicting children and adolescents. It is characterized by fusion proteins arising from chromosomal translocations. The fusion proteins induce aberrations in multiple signaling pathways and molecules, constituting a key event in oncogenic transformation. While diagnostic and therapeutic modalities have advanced in recent decades and multimodal treatments, including surgery, radiotherapy, and chemotherapy, have significantly improved survival of patients with localized tumors, patients with metastatic tumors continue to face poor prognoses. There persists a pressing need for novel alternative treatments, yet the translation of our understanding of Ewing sarcoma pathogenesis into improved clinical outcomes remains a critical challenge. Here, we provide a comprehensive review of Ewing sarcoma, including fusion proteins, various signaling pathways, pivotal pathogenetic molecules implicated in its development, and associated targeted therapies and immunotherapies. We summarize past endeavors, current advancements, and deliberate on limitations and future research directions. It is envisaged that this review will furnish novel insights into prospective treatment avenues for Ewing sarcoma.
Collapse
Affiliation(s)
- Ke Jia
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Li Cao
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Washington University School of Medicine, St Louis, MO, USA.
| | - Yihan Yu
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Doudou Jing
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| | - Wei Wu
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | | | - Zengwu Shao
- Department of Orthopaedics, Union hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
3
|
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
|
4
|
Kramer NE, Byun S, Coryell P, D’Costa S, Thulson E, Kim H, Parkus SM, Bond ML, Klein ER, Shine J, Chubinskaya S, Love MI, Mohlke KL, Diekman BO, Loeser RF, Phanstiel DH. Response eQTLs, chromatin accessibility, and 3D chromatin structure in chondrocytes provide mechanistic insight into osteoarthritis risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.05.592567. [PMID: 38952796 PMCID: PMC11216363 DOI: 10.1101/2024.05.05.592567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Osteoarthritis (OA) poses a significant healthcare burden with limited treatment options. While genome-wide association studies (GWAS) have identified over 100 OA-associated loci, translating these findings into therapeutic targets remains challenging. Integrating expression quantitative trait loci (eQTL), 3D chromatin structure, and other genomic approaches with OA GWAS data offers a promising approach to elucidate disease mechanisms; however, comprehensive eQTL maps in OA-relevant tissues and conditions remain scarce. We mapped gene expression, chromatin accessibility, and 3D chromatin structure in primary human articular chondrocytes in both resting and OA-mimicking conditions. We identified thousands of differentially expressed genes, including those associated with differences in sex and age. RNA-seq in chondrocytes from 101 donors across two conditions uncovered 3782 unique eGenes, including 420 that exhibited strong and significant condition-specific effects. Colocalization with OA GWAS signals revealed 13 putative OA risk genes, 10 of which have not been previously identified. Chromatin accessibility and 3D chromatin structure provided insights into the mechanisms and conditional specificity of these variants. Our findings shed light on OA pathogenesis and highlight potential targets for therapeutic development.
Collapse
Affiliation(s)
- Nicole E Kramer
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Seyoun Byun
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Philip Coryell
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susan D’Costa
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eliza Thulson
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - HyunAh Kim
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sylvie M Parkus
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Marielle L Bond
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Emma R Klein
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jacqueline Shine
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Susanna Chubinskaya
- Department of Pediatrics, Rush University Medical Center, Chicago, IL 60612, USA
| | - Michael I Love
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Raleigh, NC 27695, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Division of Rheumatology, Allergy and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Douglas H Phanstiel
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
5
|
Gude MF, Hjortebjerg R, Bjerre M, Pedersen AKN, Oxvig C, Rasmussen LM, Frystyk J, Steffensen L. The pro-atherogenic enzyme PAPP-A is active in eluates from human carotid and femoral atherosclerotic plaques. ATHEROSCLEROSIS PLUS 2024; 57:30-36. [PMID: 39308741 PMCID: PMC11415872 DOI: 10.1016/j.athplu.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024]
Abstract
Background Pregnancy-associated plasma protein-A (PAPP-A) regulates bioavailability of insulin-like growth factor 1 (IGF1) in various tissues by proteolytic cleavage of a subset of IGF-binding proteins (IGFBPs). Pre-clinical studies have established a role of PAPP-A in atherosclerosis and proposed that targeting the proteolytic activity of PAPP-A has therapeutic value.This study aimed to investigate whether human atherosclerotic plaques contain proteolytically active PAPP-A, a prerequisite for further considering PAPP-A as a therapeutic target in patients. Methods We obtained carotid (n = 9) and femoral (n = 11) atherosclerotic plaques from patients undergoing vascular surgery and incubated freshly harvested plaque tissue in culture media for 24 h. Subsequently, conditioned media were assayed for PAPP-A, STC2, IGFBP4, and IGF1 using immunoassays. Enzymatic activity of PAPP-A was assessed by its ability to process recombinant IGFBP4-IGF1 complexes - a specific substrate of PAPP-A - by Western blotting. Results PAPP-A and STC2 were detectable in conditioned media from both carotid and femoral plaques, with higher STC2 concentrations in eluates from carotid plaque incubations (p = 0.02). IGFBP4 and IGF1 were undetectable. Conditioned media from all 20 plaques exhibited PAPP-A proteolytic activity. However, no correlation between PAPP-A concentration and its proteolytic activity was observed, whereas the PAPP-A: STC2 molar ratio correlated with PAPP-A activity (R2 = 0.25, p = 0.03). Conclusion This study provides evidence for the presence of enzymatically active PAPP-A in atherosclerotic plaques and underscores the need for further investigating potential beneficial effects associated with targeting PAPP-A in atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Mette Faurholdt Gude
- Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rikke Hjortebjerg
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Mette Bjerre
- Medical/Steno Aarhus Research Laboratory, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Claus Oxvig
- Dept. of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA), Odense University Hospital, Odense, Denmark
- Dept. of Clinical Biochemistry, Odense University Hospital, Odense, Denmark
| | - Jan Frystyk
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Lasse Steffensen
- Dept. of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
6
|
Harboe M, Kjaer-Sorensen K, Füchtbauer EM, Fenton RA, Thomsen JS, Brüel A, Oxvig C. The metalloproteinase PAPP-A is required for IGF-dependent chondrocyte differentiation and organization. Sci Rep 2024; 14:20161. [PMID: 39215168 PMCID: PMC11364822 DOI: 10.1038/s41598-024-71062-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
Insulin-like growth factor (IGF) signaling is required for proper growth and skeletal development in vertebrates. Consequently, its dysregulation may lead to abnormalities of growth or skeletal structures. IGF is involved in the regulation of cell proliferation and differentiation of chondrocytes. However, the availability of bioactive IGF may be controlled by antagonizing IGF binding proteins (IGFBPs) in the circulation and tissues. As the metalloproteinase PAPP-A specifically cleaves members of the IGFBP family, we hypothesized that PAPP-A activity liberates bioactive IGF in cartilage. In PAPP-A knockout mice, the femur length was reduced and the mice showed a disorganized columnar organization of growth plate chondrocytes. Similarly, zebrafish lacking pappaa showed reduced length of Meckel's cartilage and disorganized chondrocytes, reminiscent of the mouse knockout phenotype. Expression of chondrocyte differentiation markers (sox9a, ihha, and col10a1) was markedly affected in Meckel's cartilage of pappaa knockout zebrafish, indicating that differentiation of chondrocytes was compromised. Additionally, the zebrafish pappaa knockout phenotype was mimicked by pharmacological inhibition of IGF signaling, and it could be rescued by treatment with exogenous recombinant IGF-I. In conclusion, our data suggests that IGF activity in the growing cartilage, and hence IGF signaling in chondrocytes, requires the presence of PAPP-A. The absence of PAPP-A causes aberrant chondrocyte organization and compromised growth in both mice and zebrafish.
Collapse
Affiliation(s)
- Mette Harboe
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus C, Denmark
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus C, Denmark
| | - Ernst-Martin Füchtbauer
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus C, Denmark
| | - Robert A Fenton
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Universitetsbyen 81, 8000, Aarhus C, Denmark.
| |
Collapse
|
7
|
Khatun M, Modhukur V, Piltonen TT, Tapanainen JS, Salumets A. Stanniocalcin Protein Expression in Female Reproductive Organs: Literature Review and Public Cancer Database Analysis. Endocrinology 2024; 165:bqae110. [PMID: 39186548 PMCID: PMC11398916 DOI: 10.1210/endocr/bqae110] [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: 06/04/2024] [Revised: 08/05/2024] [Accepted: 08/24/2024] [Indexed: 08/28/2024]
Abstract
Stanniocalcin (STC) 1 and 2 serve as antihyperglycemic polypeptide hormones with critical roles in regulating calcium and phosphate homeostasis. They additionally function as paracrine and/or autocrine factors involved in numerous physiological processes, including female reproduction. STC1 and STC2 contribute to the pathophysiology of several diseases, including female infertility- and pregnancy-associated conditions, and even tumorigenesis of reproductive organs. This comprehensive review highlights the dynamic expression patterns and potential dysregulation of STC1 and STC2, restricted to female fertility, and infertility- and pregnancy-associated diseases and conditions, such as endometriosis, polycystic ovary syndrome (PCOS), abnormal uterine bleeding, uterine polyps, and pregnancy complications, like impaired decidualization, preeclampsia, and preterm labor. Furthermore, the review elucidates the role of dysregulated STC in the progression of cancers of the reproductive system, including endometrial, cervical, and ovarian cancers. Additionally, the review evaluates the expression patterns and prognostic significance of STC in gynecological cancers by utilizing existing public datasets from The Cancer Genome Atlas to help decipher the multifaceted roles of these pleiotropic hormones in disease progression. Understanding the intricate mechanisms by which STC proteins influence all these reviewed conditions could lead to the development of targeted diagnostic and therapeutic strategies in the context of female reproductive health and oncology.
Collapse
Affiliation(s)
- Masuma Khatun
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Vijayachitra Modhukur
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
| | - Terhi T Piltonen
- Department of Obstetrics and Gynecology, Research Unit of Clinical Medicine, Medical Research Center, Oulu University Hospital, University of Oulu, 90220 Oulu, Finland
| | - Juha S Tapanainen
- Department of Obstetrics and Gynecology, University of Helsinki and Helsinki University Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland
- Department of Obstetrics and Gynaecology, HFR—Cantonal Hospital of Fribourg and University of Fribourg, 79085 Fribourg, Switzerland
| | - Andres Salumets
- Department of Obstetrics and Gynecology, Institute of Clinical Medicine, University of Tartu, 50406 Tartu, Estonia
- Competence Centre on Health Technologies, 50411 Tartu, Estonia
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet and Karolinska University Hospital, 14152 Huddinge, Stockholm, Sweden
| |
Collapse
|
8
|
Barrios V, Martín-Rivada Á, Martos-Moreno GÁ, Canelles S, Moreno-Macián F, De Mingo-Alemany C, Delvecchio M, Pajno R, Fintini D, Chowen JA, Argente J. Increased IGFBP Proteolysis, IGF-I Bioavailability, and Pappalysin Levels in Children With Prader-Willi Syndrome. J Clin Endocrinol Metab 2024; 109:e1776-e1786. [PMID: 38141219 DOI: 10.1210/clinem/dgad754] [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: 10/12/2023] [Revised: 12/18/2023] [Accepted: 12/21/2023] [Indexed: 12/25/2023]
Abstract
CONTEXT Prader-Willi syndrome (PWS) is associated with impaired growth hormone (GH) secretion and decreased insulin-like growth factor (IGF)-I levels. Pappalysins (PAPP-A, PAPP-A2) and stanniocalcins (STC-1, STC-2) regulate IGF binding-protein (IGFBP) cleavage and IGF bioavailability, but their implication in PWS is unknown. OBJECTIVE We determined serum levels of PAPP-As and STCs in association with IGF axis components in prepubertal and pubertal patients with PWS, also analyzing the effect of GH treatment. METHODS Forty children and adolescents with PWS and 120 sex- and age-matched controls were included. The effect of GH was evaluated at 6 months of treatment in 11 children. RESULTS Children with PWS had lower levels of total IGF-I, total and intact IGFBP-3, acid-labile subunit, intact IGFBP-4, and STC-1, and they had higher concentrations of free IGF-I, IGFBP-5, and PAPP-A. Patients with PWS after pubertal onset had decreased total IGF-I, total and intact IGFBP-3, and intact IGFBP-4 levels, and had increased total IGFBP-4, and STCs concentrations. GH treatment increased total IGF-I, total and intact IGFBP-3, and intact IGFBP-4, with no changes in PAPP-As, STCs, and free IGF-I levels. Standardized height correlated directly with intact IGFBP-3 and inversely with PAPP-As and the free/total IGF-I ratio. CONCLUSION The increase in PAPP-A could be involved in increased IGFBP proteolysis, promoting IGF-I bioavailability in children with PWS. Further studies are needed to establish the relationship between growth, GH resistance, and changes in the IGF axis during development and after GH treatment in these patients.
Collapse
Affiliation(s)
- Vicente Barrios
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Álvaro Martín-Rivada
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
| | - Gabriel Á Martos-Moreno
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid 28029, Spain
| | - Sandra Canelles
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Francisca Moreno-Macián
- Pediatric Endocrinology Unit, Hospital Universitario y Politécnico La Fe, Valencia 46009, Spain
| | - Carmen De Mingo-Alemany
- Pediatric Endocrinology Unit, Hospital Universitario y Politécnico La Fe, Valencia 46009, Spain
| | - Maurizio Delvecchio
- Department of Biotechnology and Applied Sciences, University of L'Aquila, Aquila 67100, Italy
| | - Roberta Pajno
- Department of Pediatrics, Endocrine Unit, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Danilo Fintini
- Ospedale Pediatrico Bambino Gesù, Prader-Willi Reference Center, Endocrinology and Diabetology Unit, IRCCS, Rome 00165, Italy
| | - Julie A Chowen
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, Madrid 28049, Spain
| | - Jesús Argente
- Departments of Pediatrics & Pediatric Endocrinology, Hospital Infantil Universitario Niño Jesús, Research Institute "La Princesa", Madrid 28009, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutriciόn (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid 28029, Spain
- IMDEA, Food Institute, CEIUAM+CSI, Cantoblanco, Madrid 28049, Spain
| |
Collapse
|
9
|
Guerra-Cantera S, Frago LM, Espinoza-Chavarria Y, Collado-Pérez R, Jiménez-Hernaiz M, Torrecilla-Parra M, Barrios V, Belsham DD, Laursen LS, Oxvig C, Argente J, Chowen JA. Palmitic Acid Modulation of the Insulin-Like Growth Factor System in Hypothalamic Astrocytes and Neurons. Neuroendocrinology 2024; 114:958-974. [PMID: 39043147 DOI: 10.1159/000540442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Insulin-like growth factor (IGF)1 and IGF2 have neuroprotective effects, but less is known regarding how other members of the IGF system, including IGF binding proteins (IGFBPs) and the regulatory proteinase pappalysin-1 (PAPP-A) and its endogenous inhibitor stanniocalcin-2 (STC2) participate in this process. Here, we analyzed whether these members of the IGF system are modified in neurons and astrocytes in response to palmitic acid (PA), a fatty acid that induces cell stress when increased centrally. METHODS Primary hypothalamic astrocyte cultures from male and female PND2 rats and the pro-opiomelanocortin (POMC) neuronal cell line, mHypoA-POMC/GFP-2, were treated with PA, IGF1 or both. To analyze the role of STC2 in astrocytes, siRNA assays were employed. RESULTS In astrocytes of both sexes, PA rapidly increased cell stress factors followed by increased Pappa and Stc2 mRNA levels and then a decrease in Igf1, Igf2, and Igfbp2 expression and cell number. Exogenous IGF1 did not revert these effects. In mHypoA-POMC/GFP-2 neurons, PA reduced cell number and Pomc and Igf1 mRNA levels, and increased Igfbp2 and Stc2, again with no effect of exogenous IGF1. PA increased STC2 expression, but no effects of decreasing its levels by interference assays or exogenous STC2 treatment in astrocytes were found. CONCLUSIONS The response of the IGF system to PA was cell and sex specific, but no protective effects of the IGFs were found. However, the modifications in hypothalamic PAPP-A and STC2 indicate that further studies are required to determine their role in the response to fatty acids and possibly in metabolic control.
Collapse
Affiliation(s)
- Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Yesenia Espinoza-Chavarria
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Roberto Collado-Pérez
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Jiménez-Hernaiz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Torrecilla-Parra
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Denise D Belsham
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Lisbeth S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
10
|
Hivert MF, White F, Allard C, James K, Majid S, Aguet F, Ardlie KG, Florez JC, Edlow AG, Bouchard L, Jacques PÉ, Karumanchi SA, Powe CE. Placental IGFBP1 levels during early pregnancy and the risk of insulin resistance and gestational diabetes. Nat Med 2024; 30:1689-1695. [PMID: 38627562 PMCID: PMC11186792 DOI: 10.1038/s41591-024-02936-5] [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: 10/18/2023] [Accepted: 03/21/2024] [Indexed: 04/30/2024]
Abstract
Reduced insulin sensitivity (insulin resistance) is a hallmark of normal physiology in late pregnancy and also underlies gestational diabetes mellitus (GDM). We conducted transcriptomic profiling of 434 human placentas and identified a positive association between insulin-like growth factor binding protein 1 gene (IGFBP1) expression in the placenta and insulin sensitivity at ~26 weeks gestation. Circulating IGFBP1 protein levels rose over the course of pregnancy and declined postpartum, which, together with high gene expression levels in our placenta samples, suggests a placental or decidual source. Higher circulating IGFBP1 levels were associated with greater insulin sensitivity (lesser insulin resistance) at ~26 weeks gestation in the same cohort and in two additional pregnancy cohorts. In addition, low circulating IGFBP1 levels in early pregnancy predicted subsequent GDM diagnosis in two cohorts of pregnant women. These results implicate IGFBP1 in the glycemic physiology of pregnancy and suggest a role for placental IGFBP1 deficiency in GDM pathogenesis.
Collapse
Affiliation(s)
- Marie-France Hivert
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA.
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, Quebec, Canada.
| | - Frédérique White
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Catherine Allard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, Quebec, Canada
| | - Kaitlyn James
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sana Majid
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | | | | | - Jose C Florez
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Luigi Bouchard
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, Quebec, Canada
- Department of Biochemistry and Functional Genomics, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Department of Medical Biology, CIUSSS of Saguenay-Lac-Saint-Jean, Saguenay, Quebec, Canada
| | - Pierre-Étienne Jacques
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke (CRCHUS), Sherbrooke, Quebec, Canada
- Département de Biologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
- Institut de Recherche sur le Cancer de l'Université de Sherbrooke (IRCUS), Sherbrooke, Quebec, Canada
| | | | - Camille E Powe
- Diabetes Unit, Endocrine Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Nimptsch K, Aydin EE, Chavarria RFR, Janke J, Poy MN, Oxvig C, Steinbrecher A, Pischon T. Pregnancy associated plasma protein-A2 (PAPP-A2) and stanniocalcin-2 (STC2) but not PAPP-A are associated with circulating total IGF-1 in a human adult population. Sci Rep 2024; 14:1770. [PMID: 38245583 PMCID: PMC10799854 DOI: 10.1038/s41598-024-52074-8] [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: 07/05/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
The pappalysins pregnancy associated plasma protein-A (PAPP-A) and -A2 (PAPP-A2) act as proteinases of insulin-like growth factor-1 (IGF-1) binding proteins, while stanniocalcin-2 (STC2) was identified as a pappalysin inhibitor. While there is some evidence from studies in children and adolescents, it is unclear whether these molecules are related to concentrations of IGF-1 and its binding proteins in adults. We investigated cross-sectionally the association of circulating PAPP-A, PAPP-A2 and STC2 with IGF-1 and its binding proteins (IGFBPs) in 394 adult pretest participants (20-69 years) of the German National Cohort Berlin North study center. Plasma PAPP-A, PAPP-A2, total and free IGF-1, IGFBP-1, IGFBP-2, IGFBP-3, IGFBP-5 and STC2 were measured by ELISAs. The associations of PAPP-A, PAPP-A2 and STC2 with IGF-1 or IGFBPs were investigated using multivariable linear regression analyses adjusting for age, sex, body mass index and pretest phase. We observed significant inverse associations of PAPP-A2 (difference in concentrations per 0.5 ng/mL higher PAPP-A2 levels) with total IGF-1 (- 4.3 ng/mL; 95% CI - 7.0; - 1.6), the IGF-1:IGFBP-3 molar ratio (- 0.34%; 95%-CI - 0.59; - 0.09), but not free IGF-1 and a positive association with IGFBP-2 (11.9 ng/mL; 95% CI 5.0; 18.8). PAPP-A was not related to total or free IGF-1, but positively associated with IGFBP-5. STC2 was inversely related to total IGF-1, IGFBP-2 and IGFBP-3 and positively to IGFBP-1. This first investigation of these associations in a general adult population supports the hypothesis that PAPP-A2 as well as STC2 play a role for IGF-1 and its binding proteins, especially for total IGF-1. The role of PAPP-A2 and STC2 for health and disease in adults warrants further investigation.
Collapse
Affiliation(s)
- Katharina Nimptsch
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany.
| | - Elif Ece Aydin
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rafael Francisco Rios Chavarria
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Jürgen Janke
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Core Facility Biobank, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Matthew N Poy
- John Hopkins University, All Children's Hospital, St. Petersburg, FL, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Astrid Steinbrecher
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany
| | - Tobias Pischon
- Molecular Epidemiology Research Group, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Straße 10, 13125, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Biobank Technology Platform, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Core Facility Biobank, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
15
|
Zafar S, Fatima SI, Schmitz M, Zerr I. Current Technologies Unraveling the Significance of Post-Translational Modifications (PTMs) as Crucial Players in Neurodegeneration. Biomolecules 2024; 14:118. [PMID: 38254718 PMCID: PMC10813409 DOI: 10.3390/biom14010118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
Neurodegenerative disorders, such as Parkinson's disease, Alzheimer's disease, and Huntington's disease, are identified and characterized by the progressive loss of neurons and neuronal dysfunction, resulting in cognitive and motor impairment. Recent research has shown the importance of PTMs, such as phosphorylation, acetylation, methylation, ubiquitination, sumoylation, nitration, truncation, O-GlcNAcylation, and hydroxylation, in the progression of neurodegenerative disorders. PTMs can alter protein structure and function, affecting protein stability, localization, interactions, and enzymatic activity. Aberrant PTMs can lead to protein misfolding and aggregation, impaired degradation, and clearance, and ultimately, to neuronal dysfunction and death. The main objective of this review is to provide an overview of the PTMs involved in neurodegeneration, their underlying mechanisms, methods to isolate PTMs, and the potential therapeutic targets for these disorders. The PTMs discussed in this article include tau phosphorylation, α-synuclein and Huntingtin ubiquitination, histone acetylation and methylation, and RNA modifications. Understanding the role of PTMs in neurodegenerative diseases may provide new therapeutic strategies for these devastating disorders.
Collapse
Affiliation(s)
- Saima Zafar
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
- Biomedical Engineering and Sciences Department, School of Mechanical and Manufacturing Engineering (SMME), National University of Sciences and Technology (NUST), Bolan Road, H-12, Islamabad 44000, Pakistan
| | - Shehzadi Irum Fatima
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| | - Matthias Schmitz
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| | - Inga Zerr
- Department of Neurology, Clinical Dementia Center and DZNE, University Medical Center Goettingen (UMG), Georg-August University, Robert-Koch-Str. 40, 37075 Goettingen, Germany
| |
Collapse
|
16
|
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
|
17
|
Li S, Li H, Wang Z, Duan C. Stanniocalcin 1a regulates organismal calcium balance and survival by suppressing Trpv6 expression and inhibiting IGF signaling in zebrafish. Front Endocrinol (Lausanne) 2023; 14:1276348. [PMID: 37964974 PMCID: PMC10640984 DOI: 10.3389/fendo.2023.1276348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
Stanniocalcin 1 (Stc1) is well known for its role in regulating calcium uptake in fish by acting on ionocytes or NaR cells. A hallmark of NaR cells is the expression of Trpv6, a constitutively open calcium channel. Recent studies in zebrafish suggest that genetical deletion of Stc1a and Trpv6 individually both increases IGF signaling and NaR cell proliferation. While trpv6-/- fish suffered from calcium deficiency and died prematurely, stc1a-/- fish had elevated body calcium levels but also died prematurely. The relationship between Stc1a, Trpv6, and IGF signaling in regulating calcium homeostasis and organismal survival is unclear. Here we report that loss of Stc1a increases Trpv6 expression in NaR cells in an IGF signaling-dependent manner. Treatment with CdCl2, a Trpv6 inhibitor, reduced NaR cell number in stc1a -/- fish to the sibling levels. Genetic and biochemical analysis results suggest that Stc1a and Trpv6 regulate NaR cell proliferation via the same IGF pathway. Alizarin red staining detected abnormal calcium deposits in the yolk sac region and kidney stone-like structures in stc1a -/- fish. Double knockout or pharmacological inhibition of Trpv6 alleviated these phenotypes, suggesting that Stc1a inhibit epithelial Ca2+ uptake by regulating Trpv6 expression and activity. stc1a-/- mutant fish developed cardiac edema, body swelling, and died prematurely. Treatment of stc1a-/- fish with CdCl2 or double knockout of Trpv6 alleviated these phenotypes. These results provide evidence that Stc1a regulates calcium homeostasis and organismal survival by suppressing Trpv6 expression and inhibiting IGF signaling in ionocytes.
Collapse
Affiliation(s)
| | | | | | - Cunming Duan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
18
|
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
|
19
|
Hjortebjerg R, Pedersen DA, Mengel-From J, Jørgensen LH, Christensen K, Frystyk J. Heritability and circulating concentrations of pregnancy-associated plasma protein-A and stanniocalcin-2 in elderly monozygotic and dizygotic twins. Front Endocrinol (Lausanne) 2023; 14:1193742. [PMID: 37334305 PMCID: PMC10272750 DOI: 10.3389/fendo.2023.1193742] [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: 03/25/2023] [Accepted: 05/15/2023] [Indexed: 06/20/2023] Open
Abstract
Introduction Pregnancy-associated plasma protein-A (PAPP-A) is an IGF-activating enzyme suggested to influence aging-related diseases. However, knowledge on serum PAPP-A concentration and regulation in elderly subjects is limited. Therefore, we measured serum PAPP-A in elderly same-sex monozygotic (MZ) and dizygotic (DZ) twins, as this allowed us to describe the age-relationship of PAPP-A, and to test the hypothesis that serum PAPP-A concentrations are genetically determined. As PAPP-A is functionally related to stanniocalcin-2 (STC2), an endogenous PAPP-A inhibitor, we included measurements on STC2 as well as IGF-I and IGF-II. Methods The twin cohort contained 596 subjects (250 MZ twins, 346 DZ twins), whereof 33% were males. The age ranged from 73.2 to 94.3 (mean 78.8) years. Serum was analyzed for PAPP-A, STC2, IGF-I, and IGF-II by commercial immunoassays. Results In the twin cohort, PAPP-A increased with age (r=0.19; P<0.05), whereas IGF-I decreased (r=-0.12; P<0.05). Neither STC2 nor IGF-II showed any age relationship. When analyzed according to sex, PAPP-A correlated positively with age in males (r=0.18; P<0.05) and females (r=0.25; P<0.01), whereas IGF-I correlated inversely in females only (r=-0.15; P<0.01). Males had higher levels of PAPP-A (29%), STC2 (18%) and IGF-I (19%), whereas serum IGF-II was 28% higher in females (all P<0.001). For all four proteins, within-pair correlations were significantly higher for MZ twins than for DZ twins, and they demonstrated substantial and significant heritability, which after adjustment for age and sex averaged 59% for PAPP-A, 66% for STC2, 58% for IGF-I, and 52% for IGF-II. Discussion This twin study confirms our hypothesis that the heritability of PAPP-A serum concentrations is substantial, and the same is true for STC2. As regards the age relationship, PAPP-A increases with age, whereas STC2 remains unchanged, thereby supporting the idea that the ability of STC2 to inhibit PAPP-A enzymatic activity decreases with increasing age.
Collapse
Affiliation(s)
- Rikke Hjortebjerg
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
| | - Dorthe Almind Pedersen
- The Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
| | - Jonas Mengel-From
- The Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | | | - Kaare Christensen
- The Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Jan Frystyk
- Endocrine Research Unit, Department of Endocrinology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
20
|
Guibourdenche J, Leguy MC, Pidoux G, Hebert-Schuster M, Laguillier C, Anselem O, Grangé G, Bonnet F, Tsatsaris V. Biochemical Screening for Fetal Trisomy 21: Pathophysiology of Maternal Serum Markers and Involvement of the Placenta. Int J Mol Sci 2023; 24:ijms24087669. [PMID: 37108840 PMCID: PMC10146970 DOI: 10.3390/ijms24087669] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
It is now well established that maternal serum markers are often abnormal in fetal trisomy 21. Their determination is recommended for prenatal screening and pregnancy follow-up. However, mechanisms leading to abnormal maternal serum levels of such markers are still debated. Our objective was to help clinicians and scientists unravel the pathophysiology of these markers via a review of the main studies published in this field, both in vivo and in vitro, focusing on the six most widely used markers (hCG, its free subunit hCGβ, PAPP-A, AFP, uE3, and inhibin A) as well as cell-free feto-placental DNA. Analysis of the literature shows that mechanisms underlying each marker's regulation are multiple and not necessarily directly linked with the supernumerary chromosome 21. The crucial involvement of the placenta is also highlighted, which could be defective in one or several of its functions (turnover and apoptosis, endocrine production, and feto-maternal exchanges and transfer). These defects were neither constant nor specific for trisomy 21, and might be more or less pronounced, reflecting a high variability in placental immaturity and alteration. This explains why maternal serum markers can lack both specificity and sensitivity, and are thus restricted to screening.
Collapse
Affiliation(s)
- Jean Guibourdenche
- Hormonologie CHU Cochin AP-HP, 75014 Paris, France
- Faculté de Santé, Université Paris Cité, 75014 Paris, France
- FHU Préma, 75014 Paris, France
| | | | | | | | - Christelle Laguillier
- Hormonologie CHU Cochin AP-HP, 75014 Paris, France
- Faculté de Santé, Université Paris Cité, 75014 Paris, France
- UMR-S1139, 75014 Paris, France
| | - Olivia Anselem
- FHU Préma, 75014 Paris, France
- Maternité Port Royal CHU Cochin AP-HP, 75014 Paris, France
| | - Gilles Grangé
- FHU Préma, 75014 Paris, France
- Maternité Port Royal CHU Cochin AP-HP, 75014 Paris, France
| | - Fidéline Bonnet
- Hormonologie CHU Cochin AP-HP, 75014 Paris, France
- Faculté de Santé, Université Paris Cité, 75014 Paris, France
| | - Vassilis Tsatsaris
- Faculté de Santé, Université Paris Cité, 75014 Paris, France
- FHU Préma, 75014 Paris, France
- Maternité Port Royal CHU Cochin AP-HP, 75014 Paris, France
| |
Collapse
|