1
|
Flores-Espinosa P, Menon R, Kammala A, Richardson LS. Lead exposure at the feto-maternal interface: a cause for concern for fetal membrane trophoblasts. Toxicol Sci 2025; 203:195-205. [PMID: 39579145 PMCID: PMC11775422 DOI: 10.1093/toxsci/kfae149] [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] [Indexed: 11/25/2024] Open
Abstract
The integrity of fetal membranes enables biological functions that protect the fetus and maintain the pregnancy. Any compromise in fetal membrane function can predispose a pregnant woman to prelabor rupture of the membranes (pPROMs) and subsequently to preterm birth (PTB). Epidemiologic data suggest that lead exposure during pregnancy is one of several risk factors associated with PTB and pPROM. This heavy metal can cross placental and fetal membrane barriers, disrupting homeostasis in these tissues. Autophagy contributes to the maintenance of fetal membrane homeostasis during gestation, and dysfunctional autophagy is associated with pPROM. In this study, we determined the mechanistic impact of lead-induced cellular changes, autophagy, senescence, and inflammation in chorion trophoblast cells (CTCs) and amnion epithelial cells (AECs) of the fetal membranes. Lead exposure in CTCs induced autophagy dysfunction (increase in LC3B-II), augmented senescence (increased SA-β-galactosidase activity), and increased the release of inflammation. In AECs, lead exposure did effect autophagy, senescence, nor inflammation. The differential changes observed in CTCs and AECs after exposure to high lead concentrations may promote the weakening of fetal membranes and contribute to preterm rupture.
Collapse
Affiliation(s)
- Pilar Flores-Espinosa
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, United States
- Laboratorio de Inmunología de la Unidad Feto-Placentaria, Department of Immunobiochemestry, Instituto Nacional de Perinatología I.E.R, Mexico City 11000, Mexico
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, United States
| | - Ananth Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, United States
| | - Lauren S Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX 77555-1062, United States
| |
Collapse
|
2
|
Costa E, Thrasivoulou C, Becker DL, Deprest J, David AL, Chowdhury TT. Role of Myofibroblasts in the Repair of Iatrogenic Preterm Membranes Subjected to Mechanical Stimulation. Prenat Diagn 2025; 45:102-112. [PMID: 39631799 PMCID: PMC11717736 DOI: 10.1002/pd.6722] [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: 03/15/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
OBJECTIVE We examined the role of myofibroblasts in regulating Cx43 and collagen structure in iatrogenic preterm amniotic membrane (AM) defects subjected to mechanical stimulation. METHOD Preterm AM specimens were collected from women undergoing planned preterm caesarean section after in utero intervention for correction of spina bifida by open fetal surgery (n = 4 patients; preterm delivery at 34 + 0 weeks to 35 + 0 weeks). Control specimens taken 5 cm away from the open fetal surgery defect site were compared with wound edge AM. In separate experiments, the effects of mechanical stimulation and co-treatment with Cx43 antisense on matrix and repair proteins were examined. Specimens were immunostained to detect αSMA and Cx43 in myofibroblasts and counterstained with DAPI to quantify nuclei shape. The direction of collagen fibrils in the wound edge region was examined by SHG imaging. Markers for matrix (collagen, elastin, GAG), inflammation (PGE2) and repair (TGFβ1) were examined by RT-qPCR and biochemical assays. RESULTS In iatrogenic preterm AM specimens, the diameter of the open fetal surgery defect ranged between 3.5 and 7.5 cm. At the wound edge of the open fetal surgery defect, αSMA positive myofibroblasts had deformed nuclei and showed abundant Cx43 localized in the cell bodies or formed plaques. In the fibroblast layer, collagen had degenerated in some regions or had polarity near the wound edge. In preterm AM defects, mechanical stimulation and Cx43 antisense increased the levels of collagen and elastin but not GAG or PGE2 release. Mechanical stimulation increased Cx43 and TGFβ1 gene expression. CONCLUSION In open fetal surgery defects, myofibroblasts were elongated with collagen fibrils that either degenerated or had polarity. Whilst cells produced substantially higher Cx43 in the fibroblast than in the epithelial layer, they formed plaques, which may prevent migration and delay healing. Mechanical stimulation of preterm AM enhanced matrix repair proteins and the mechanotransduction should be explored to understand how Cx43 contributes to membrane integrity.
Collapse
Affiliation(s)
- E. Costa
- Centre for BioengineeringSchool of Engineering and Materials ScienceQueen Mary University of LondonLondonUK
| | - C. Thrasivoulou
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - D. L. Becker
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingaporeSingapore
| | - J. Deprest
- Department of Obstetrics and GynecologyUniversity Hospitals LeuvenLeuvenBelgium
- EGA Institute for Women's HealthUniversity College LondonLondonUK
| | - A. L. David
- EGA Institute for Women's HealthUniversity College LondonLondonUK
| | - T. T. Chowdhury
- Centre for BioengineeringSchool of Engineering and Materials ScienceQueen Mary University of LondonLondonUK
| |
Collapse
|
3
|
Prencipe G, Cerveró-Varona A, Perugini M, Sulcanese L, Iannetta A, Haidar-Montes AA, Stöckl J, Canciello A, Berardinelli P, Russo V, Barboni B. Amphiregulin orchestrates the paracrine immune-suppressive function of amniotic-derived cells through its interplay with COX-2/PGE 2/EP4 axis. iScience 2024; 27:110508. [PMID: 39156643 PMCID: PMC11326934 DOI: 10.1016/j.isci.2024.110508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/10/2024] [Accepted: 07/11/2024] [Indexed: 08/20/2024] Open
Abstract
The paracrine crosstalk between amniotic-derived membranes (AMs)/epithelial cells (AECs) and immune cells is pivotal in tissue healing following inflammation. Despite evidence collected to date, gaps in understanding the underlying molecular mechanisms have hindered clinical applications. The present study represents a significant step forward demonstrating that amphiregulin (AREG) orchestrates the native immunomodulatory functions of amniotic derivatives via the COX-2/PGE2/EP4 axis. The results highlight the immunosuppressive efficacy of PGE2-dependent AREG release, dampening PBMCs' activation, and NFAT pathway in Jurkat reporter cells via TGF-β signaling. Moreover, AREG emerges as a key protein mediator by attenuating acute inflammatory response in Tg(lysC:DsRed2) zebrafish larvae. Notably, the interplay of diverse COX-2/PGE2 pathway activators enables AM/AEC to adapt rapidly to external stimuli (LPS and/or stretching) through a responsive positive feedback loop on the AREG/EGFR axis. These findings offer valuable insights for developing innovative cell-free therapies leveraging the potential of amniotic derivatives in immune-mediated diseases and regenerative medicine.
Collapse
Affiliation(s)
- Giuseppe Prencipe
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Adrián Cerveró-Varona
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Ludovica Sulcanese
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Annamaria Iannetta
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Arlette Alina Haidar-Montes
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Johannes Stöckl
- Centre for Pathophysiology, Infectiology and Immunology, Institute of Immunology, Medical University of Vienna, Vienna 1090, Austria
| | - Angelo Canciello
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Paolo Berardinelli
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Valentina Russo
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| | - Barbara Barboni
- Unit of Basic and Applied Sciences, Department of Biosciences and Agro-Food and Environmental Technologies, University of Teramo, 64100 Teramo, Italy
| |
Collapse
|
4
|
Padron JG, Saito Reis CA, Ng PK, Norman Ing ND, Baker H, Davis K, Kurashima C, Kendal-Wright CE. Stretch Causes cffDNA and HMGB1-Mediated Inflammation and Cellular Stress in Human Fetal Membranes. Int J Mol Sci 2024; 25:5161. [PMID: 38791199 PMCID: PMC11121497 DOI: 10.3390/ijms25105161] [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: 03/26/2024] [Revised: 04/27/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Danger-associated molecular patterns (DAMPs) are elevated within the amniotic cavity, and their increases correlate with advancing gestational age, chorioamnionitis, and labor. Although the specific triggers for their release in utero remain unclear, it is thought that they may contribute to the initiation of parturition by influencing cellular stress mechanisms that make the fetal membranes (FMs) more susceptible to rupture. DAMPs induce inflammation in many different tissue types. Indeed, they precipitate the subsequent release of several proinflammatory cytokines that are known to be key for the weakening of FMs. Previously, we have shown that in vitro stretch of human amnion epithelial cells (hAECs) induces a cellular stress response that increases high-mobility group box-1 (HMGB1) secretion. We have also shown that cell-free fetal DNA (cffDNA) induces a cytokine response in FM explants that is fetal sex-specific. Therefore, the aim of this work was to further investigate the link between stretch and the DAMPs HMGB1 and cffDNA in the FM. These data show that stretch increases the level of cffDNA released from hAECs. It also confirms the importance of the sex of the fetus by demonstrating that female cffDNA induced more cellular stress than male fetuses. Our data treating hAECs and human amnion mesenchymal cells with HMGB1 show that it has a differential effect on the ability of the cells of the amnion to upregulate the proinflammatory cytokines and propagate a proinflammatory signal through the FM that may weaken it. Finally, our data show that sulforaphane (SFN), a potent activator of Nrf2, is able to mitigate the proinflammatory effects of stretch by decreasing the levels of HMGB1 release and ROS generation after stretch and modulating the increase of key cytokines after cell stress. HMGB1 and cffDNA are two of the few DAMPs that are known to induce cytokine release and matrix metalloproteinase (MMP) activation in the FMs; thus, these data support the general thesis that they can function as potential central players in the normal mechanisms of FM weakening during the normal distension of this tissue at the end of a normal pregnancy.
Collapse
Affiliation(s)
- Justin Gary Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96822, USA;
- Wayne State School of Medicine, Detroit, MI 48201, USA
| | - Chelsea A. Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Po’okela K. Ng
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Nainoa D. Norman Ing
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Hannah Baker
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Kamalei Davis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Courtney Kurashima
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
| | - Claire E. Kendal-Wright
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96822, USA;
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (C.A.S.R.); (P.K.N.); (N.D.N.I.); (H.B.); (K.D.); (C.K.)
- Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96826, USA
| |
Collapse
|
5
|
Costa E, Thrasivoulou C, Becker DL, Deprest JA, David AL, Chowdhury TT. Cx43 regulates mechanotransduction mechanisms in human preterm amniotic membrane defects. Prenat Diagn 2023; 43:1284-1295. [PMID: 37649228 DOI: 10.1002/pd.6429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
OBJECTIVE The effects of mechanical stimulation in preterm amniotic membrane (AM) defects were explored. METHODS Preterm AM was collected from women undergoing planned preterm caesarean section (CS) due to fetal growth restriction or emergency CS after spontaneous preterm prelabour rupture of the membranes (sPPROM). AM explants near the cervix or placenta were subjected to trauma and/or mechanical stimulation with the Cx43 antisense. Markers for nuclear morphology (DAPI), myofibroblasts (αSMA), migration (Cx43), inflammation (PGE2 ) and repair (collagen, elastin and transforming growth factor β [TGFβ1 ]) were examined by confocal microscopy, second harmonic generation, qPCR and biochemical assays. RESULTS In preterm AM defects, myofibroblast nuclei were highly deformed and contractile and expressed αSMA and Cx43. Mechanical stimulation increased collagen fibre polarisation and the effects on matrix markers were dependent on tissue region, disease state, gestational age and the number of fetuses. PGE2 levels were broadly similar but reduced after co-treatment with Cx43 antisense in late sPPROM AM defects. TGFβ1 and Cx43 gene expression were significantly increased after trauma and mechanical stimulation but this response dependent on gestational age. CONCLUSION Mechanical stimulation affects Cx43 signalling and cell/collagen mechanics in preterm AM defects. Establishing how Cx43 regulates mechanosignalling could be an approach to repair tissue integrity after trauma.
Collapse
Affiliation(s)
- Eleni Costa
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | | | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jan A Deprest
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, Medical School Building, London, UK
| | - Anna L David
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, Medical School Building, London, UK
| | - Tina T Chowdhury
- Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| |
Collapse
|
6
|
Vidal MS, Lintao RCV, Severino MEL, Tantengco OAG, Menon R. Spontaneous preterm birth: Involvement of multiple feto-maternal tissues and organ systems, differing mechanisms, and pathways. Front Endocrinol (Lausanne) 2022; 13:1015622. [PMID: 36313741 PMCID: PMC9606232 DOI: 10.3389/fendo.2022.1015622] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Survivors of preterm birth struggle with multitudes of disabilities due to improper in utero programming of various tissues and organ systems contributing to adult-onset diseases at a very early stage of their lives. Therefore, the persistent rates of low birth weight (birth weight < 2,500 grams), as well as rates of neonatal and maternal morbidities and mortalities, need to be addressed. Active research throughout the years has provided us with multiple theories regarding the risk factors, initiators, biomarkers, and clinical manifestations of spontaneous preterm birth. Fetal organs, like the placenta and fetal membranes, and maternal tissues and organs, like the decidua, myometrium, and cervix, have all been shown to uniquely respond to specific exogenous or endogenous risk factors. These uniquely contribute to dynamic changes at the molecular and cellular levels to effect preterm labor pathways leading to delivery. Multiple intervention targets in these different tissues and organs have been successfully tested in preclinical trials to reduce the individual impacts on promoting preterm birth. However, these preclinical trial data have not been effectively translated into developing biomarkers of high-risk individuals for an early diagnosis of the disease. This becomes more evident when examining the current global rate of preterm birth, which remains staggeringly high despite years of research. We postulate that studying each tissue and organ in silos, as how the majority of research has been conducted in the past years, is unlikely to address the network interaction between various systems leading to a synchronized activity during either term or preterm labor and delivery. To address current limitations, this review proposes an integrated approach to studying various tissues and organs involved in the maintenance of normal pregnancy, promotion of normal parturition, and more importantly, contributions towards preterm birth. We also stress the need for biological models that allows for concomitant observation and analysis of interactions, rather than focusing on these tissues and organ in silos.
Collapse
Affiliation(s)
- Manuel S. Vidal
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ryan C. V. Lintao
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Mary Elise L. Severino
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ourlad Alzeus G. Tantengco
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
7
|
Richardson L, Menon R. Fetal membrane at the feto-maternal interface: An underappreciated and understudied intrauterine tissue. PLACENTA AND REPRODUCTIVE MEDICINE 2022; 1:10.54844/prm.2022.0104. [PMID: 37502422 PMCID: PMC10373051 DOI: 10.54844/prm.2022.0104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Affiliation(s)
- Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555, TX, USA
| |
Collapse
|
8
|
Saito Reis CA, Ng PK, Kurashima CK, Padron J, Kendal-Wright CE. Fetal DNA Causes Sex-Specific Inflammation From Human Fetal Membranes. Front Physiol 2022; 13:901726. [PMID: 35812324 PMCID: PMC9257279 DOI: 10.3389/fphys.2022.901726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Inflammation is central to the mechanisms of parturition, but the lack of understanding of how it is controlled in normal parturition hampers our ability to understand how it may diverge resulting in preterm birth. Cell-free fetal DNA is found in the amniotic fluid, and it is thought to be able to activate inflammation as a danger-associated molecular pattern. Although its levels increases with gestational age, its effect has not been studied on the human fetal membranes. Thus, the aim of this study was to determine if the fetal DNA can trigger inflammation in the human fetal membranes and, thus, potentially contribute to the inflammatory load. Isolated human amniotic epithelial cells and fetal membrane explants were treated apically with fetal DNA causing the translocation of NF-KB into the nucleus of cells and throughout the cells of the explant layers with time. Fetal membrane explants were treated apically with either small or larger fragments of fetal DNA. IL-6, TNFα, and GM-CSF secretion was measured by ELISA, and pro-MMP2 and pro-MMP9 activity was measured by zymography from apical and basal media. Increased apical IL-6 secretion and basal pro-MMP2 activity was seen with small fragments of fetal DNA. When the data were disaggregated based on fetal sex, males had significant increases in IL-6 secretion and basal increased activity in pro-MMP2 and 9, whereas females had significantly increased basal secretion of TNFα. This was caused by the smaller fragments of fetal DNA, whereas the larger fragments did not cause any significant increases. Male fetal DNA had significantly lower percentages of methylation than females. Thus, when the cytokine and pro-MMP activity data were correlated with methylation percentage, IL-6 secretion significantly correlated negatively, whereas GM-CSF secretion positively correlated. These data support the role of fetal DNA as an inflammatory stimulus in the FM, as measured by increased NF-κB translocation, cytokine secretion, and increased pro-MMP activity. However, the data also suggested that the responses are different from FM tissues of male and female fetuses, and both the fragment size and methylation status of the fetal DNA can influence the magnitude and type of molecule secreted.
Collapse
Affiliation(s)
- Chelsea A. Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | - Po’okela K. Ng
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | | | - Justin Padron
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Claire Enid Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
- Department of Obstetrics, Gynecology and Women’s Health, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
- *Correspondence: Claire Enid Kendal-Wright,
| |
Collapse
|
9
|
Bhunia S, O'Brien S, Ling Y, Huang Z, Wu P, Yang Y. New approaches suggest term and preterm human fetal membranes may have distinct biomechanical properties. Sci Rep 2022; 12:5109. [PMID: 35332209 PMCID: PMC8948223 DOI: 10.1038/s41598-022-09005-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 03/04/2022] [Indexed: 12/28/2022] Open
Abstract
Preterm prelabour rupture of membranes is the leading cause of preterm birth and its associated infant mortality and morbidity. However, its underlying mechanism remains unknown. We utilized two novel biomechanical assessment techniques, ball indentation and Optical Coherence Elastography (OCE), to compare the mechanical properties and behaviours of term (≥ 37 weeks) and preterm (33-36 weeks) human fetal membranes from ruptured and non-ruptured regions. We defined the expression levels of collagen, sulfated glycosaminoglycans (sGAG), matrix metalloproteinase (MMP-9, MMP-13), fibronectin, and interleukin-1β (IL-1β) within membranes by biochemical analysis, immunohistochemical staining and Western blotting, both with and without simulated fetal movement forces on membrane rupture with a new loading system. Preterm membranes showed greater heterogeneity in mechanical properties/behaviours between ruptured and non-ruptured regions compared with their term counterparts (displacement rate: 36% vs. 15%; modulus: 125% vs. 34%; thickness: 93% vs. 30%; collagen content: 98% vs. 29%; sGAG: 85% vs 25%). Furthermore, simulated fetal movement forces triggered higher MMP-9, MMP-13 and IL-1β expression in preterm than term membranes, while nifedipine attenuated the observed increases in expression. In conclusion, the distinct biomechanical profiles of term and preterm membranes and the abnormal biochemical expression and activation by external forces in preterm membranes may provide insights into mechanisms of preterm rupture of membranes.
Collapse
Affiliation(s)
- Sudeshna Bhunia
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK
| | - Shaughn O'Brien
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK.,Academic Department of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, ST4 6QG, UK
| | - Yuting Ling
- School of Science and Engineering, University of Dundee, Dundee, DD1 4HN, UK
| | - Zhihong Huang
- School of Science and Engineering, University of Dundee, Dundee, DD1 4HN, UK
| | - Pensée Wu
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK. .,Academic Department of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, ST4 6QG, UK. .,School of Medicine, Keele University, Staffordshire, ST5 5BG, UK.
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, ST4 7QB, UK.
| |
Collapse
|
10
|
Belville C, Ponelle-Chachuat F, Rouzaire M, Gross C, Pereira B, Gallot D, Sapin V, Blanchon L. Physiological TLR4 regulation in human fetal membranes as an explicative mechanism of a pathological preterm case. eLife 2022; 11:71521. [PMID: 35119365 PMCID: PMC8816379 DOI: 10.7554/elife.71521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022] Open
Abstract
The integrity of human fetal membranes is crucial for harmonious fetal development throughout pregnancy. Their premature rupture is often the consequence of a physiological phenomenon that has been exacerbated. Beyond all the implied biological processes, inflammation is of primary importance and is qualified as ‘sterile’ at the end of pregnancy. In this study, complementary methylomic and transcriptomic strategies on amnion and choriodecidua explants obtained from the altered (cervix zone) and intact fetal membranes at term and before labour were used. By cross-analysing genome-wide studies strengthened by in vitro experiments, we deciphered how the expression of toll-like receptor 4 (TLR4), an actor in pathological fetal membrane rupture, is controlled. Indeed, it is differentially regulated in the altered zone and between both layers by a dual mechanism: (1) the methylation of TLR4 and miRNA promoters and (2) targeting by miRNA (let-7a-2 and miR-125b-1) acting on the 3’-UTR of TLR4. Consequently, this study demonstrates that fine regulation of TLR4 is required for sterile inflammation establishment at the end of pregnancy and that it may be dysregulated in the pathological premature rupture of membranes.
Collapse
Affiliation(s)
- Corinne Belville
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France
| | - Flora Ponelle-Chachuat
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France
| | - Marion Rouzaire
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France
| | - Christelle Gross
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France
| | - Bruno Pereira
- CHU Clermont-Ferrand, Biostatistics unit (DRCI) Department, clermont-ferrand, France
| | - Denis Gallot
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France.,CHU Clermont-Ferrand, Obstetrics and Gynaecology Department, Clermont-ferrand, France
| | - Vincent Sapin
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France.,CHU Clermont-Ferrand, Biochemistry and Molecular Genetic Department, Clermont-Ferrand, France
| | - Loïc Blanchon
- Team 'Translational approach to epithelial injury and repair', iGReD, Université Clermont Auvergne, Clermont-ferrand, France
| |
Collapse
|
11
|
Radnaa E, Urrabaz-Garza R, Elrod ND, de Castro Silva M, Pyles R, Han A, Menon R. Generation and characterization of human Fetal membrane and Decidual cell lines for reproductive biology experiments†. Biol Reprod 2021; 106:568-582. [PMID: 34935931 PMCID: PMC8934701 DOI: 10.1093/biolre/ioab231] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 12/10/2021] [Indexed: 12/24/2022] Open
Abstract
Human fetal membrane and maternal decidua parietalis form one of the major feto-maternal interfaces during pregnancy. Studies on this feto-maternal interface is limited as several investigators have limited access to the placenta, and experience difficulties to isolate and maintain primary cells. Many cell lines that are currently available do not have the characteristics or properties of their primary cells of origin. Therefore, we created, characterized the immortalized cells from primary isolates from fetal membrane-derived amnion epithelial cells, amnion and chorion mesenchymal cells, chorion trophoblast cells and maternal decidua parietalis cells. Primary cells were isolated from a healthy full-term, not in labor placenta. Primary cells were immortalized using either a HPV16E6E7 retroviral or a SV40T lentiviral system. The immortalized cells were characterized for the morphology, cell type-specific markers, and cell signalling pathway activation. Genomic stability of these cells was tested using RNA seq, karyotyping, and short tandem repeats DNA analysis. Immortalized cells show their characteristic morphology, and express respective epithelial, mesenchymal and decidual markers similar to that of primary cells. Gene expression of immortalized and primary cells were highly correlated (R = 0.798 to R = 0.974). Short tandem repeats DNA analysis showed in the late passage number (>P30) of cell lines matched 84-100% to the early passage number (<P10) of the cell lines revealing there were no genetic drift over the passages. Karyotyping also revealed no chromosomal anomalies. Creation of these cell lines can standardize experimental approaches, eliminate subject to subject variabilities, and benefit the reproductive biological studies on pregnancies by using these cells.
Collapse
Affiliation(s)
- Enkhtuya Radnaa
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynaecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Rheanna Urrabaz-Garza
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynaecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Nathan D Elrod
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555-0144, USA
| | - Mariana de Castro Silva
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynaecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Richard Pyles
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555-0144, USA
| | - Arum Han
- Department of Electrical and Computer Engineering, Department of Biomedical Engineering, Texas A&M University, College Station, Texas 77843-3128, USA
| | - Ramkumar Menon
- Correspondence: Department of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| |
Collapse
|
12
|
Leimert KB, Xu W, Princ MM, Chemtob S, Olson DM. Inflammatory Amplification: A Central Tenet of Uterine Transition for Labor. Front Cell Infect Microbiol 2021; 11:660983. [PMID: 34490133 PMCID: PMC8417473 DOI: 10.3389/fcimb.2021.660983] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/30/2021] [Indexed: 11/23/2022] Open
Abstract
In preparation for delivery, the uterus transitions from actively maintaining quiescence during pregnancy to an active parturient state. This transition occurs as a result of the accumulation of pro-inflammatory signals which are amplified by positive feedback interactions involving paracrine and autocrine signaling at the level of each intrauterine cell and tissue. The amplification events occur in parallel until they reach a certain threshold, ‘tipping the scale’ and contributing to processes of uterine activation and functional progesterone withdrawal. The described signaling interactions all occur upstream from the presentation of clinical labor symptoms. In this review, we will: 1) describe the different physiological processes involved in uterine transition for each intrauterine tissue; 2) compare and contrast the current models of labor initiation; 3) introduce innovative models for measuring paracrine inflammatory interactions; and 4) discuss the therapeutic value in identifying and targeting key players in this crucial event for preterm birth.
Collapse
Affiliation(s)
- Kelycia B Leimert
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Magdalena M Princ
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
13
|
Monjezi M, Jamaati H, Noorbakhsh F. Attenuation of ventilator-induced lung injury through suppressing the pro-inflammatory signaling pathways: A review on preclinical studies. Mol Immunol 2021; 135:127-136. [PMID: 33895577 DOI: 10.1016/j.molimm.2021.04.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/28/2021] [Accepted: 04/06/2021] [Indexed: 10/21/2022]
Abstract
Mechanical ventilation (MV) is a relatively common medical intervention in ICU patients. The main side effect of MV is the so-called "ventilator-induced lung injury" (VILI). The pathogenesis of VILI is not completely understood; however, it has been reported that MV might be associated with up-regulation of various inflammatory mediators within the lung tissue and that these mediators might act as pathogenic factors in lung tissue injury. One potential mechanism for the generation of inflammatory mediators is through the release of endogenous molecules known as damage associated molecular patterns (DAMPs). These molecules are released from injured tissues and can bind to pattern recognition receptors (PRRs). PRR activation generally leads to the production and release of inflammation-related molecules including innate immune cytokines and chemokines. It has been suggested that blocking DAMP/PRR signaling pathways might diminish the progression of VILI. Herein, we review the latest findings with regard to the effects of DAMP/PRRs and their blockade, as well as the potential therapeutic targets and future research directions in VILI. Results of studies performed on human samples, animal models of disease, as well as relevant in vitro systems will be discussed.
Collapse
Affiliation(s)
- Mojdeh Monjezi
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamidreza Jamaati
- Chronic Respiratory Diseases Research Center (CRDRC), National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farshid Noorbakhsh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Richardson LS, Radnaa E, Urrabaz-Garza R, Lavu N, Menon R. Stretch, scratch, and stress: Suppressors and supporters of senescence in human fetal membranes. Placenta 2020; 99:27-34. [PMID: 32750642 PMCID: PMC7530028 DOI: 10.1016/j.placenta.2020.07.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/14/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Throughout gestation, amnion membranes undergo mechanical and or physiological stretch, scratch, or stress which is withstood by repairing and remodeling processes to protect the growing fetus. At term, increased oxidative stress (OS) activates p38MAPK, induces senescence, and inflammation contributing to membrane dysfunction to promote labor. However, the signaling initiated by stretch and scratch is still unclear. This study compares the induction of p38MAPK mediated senescence by stretch, scratch, and stress in human amnion epithelial cells (AECs). METHODS Primary AECs from term, not-in-labor, fetal membranes were cultured using the following conditions (N = 3); 1) CellFlex chambers with or without 20% biaxial stretch, 2) 8-well coverslips with or without scratch, and 3) cells exposed to cigarette smoke extract (CSE) inducing OS. p38MAPK (Western blot or immunocytochemistry), senescence activation, and inflammation (matrix metalloproteinases 9 [MMP9] activity-ELISA) were determined in cells exposed to various conditions. T-test and One-Way ANOVA was used to assess significance. RESULTS Biological membrane extension, mimicked by 20% biaxial stretch of AEC, maintained an epithelial morphology and activated P-p38MAPK (P = 0.02) compared to the non-stretch controls, but did not induce senescence or MMP9 activation. AEC scratches were healed within 40-hrs, which included proliferation, migration, and cellular transitions aided by p38MAPK activation but not senescence. CSE induced OS increased p38MAPK (P = 0.018) activation, senescence (P = 0.019), and MMP9 (P = 0.02). CONCLUSION Physiologic stretch and scratch experienced during gestation can cause p38MAPK activation without causing senescence or inflammation. This may be indicative of p38MAPK's role in tissue remodeling during pregnancy. Overwhelming OS, experienced at term, results in P-p38MAPK mediated senescence and inflammation to disrupt membrane remodeling.
Collapse
Affiliation(s)
- Lauren S Richardson
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Enkhtuya Radnaa
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Rheanna Urrabaz-Garza
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Narmada Lavu
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| |
Collapse
|
15
|
Menon R, Behnia F, Polettini J, Richardson LS. Novel pathways of inflammation in human fetal membranes associated with preterm birth and preterm pre-labor rupture of the membranes. Semin Immunopathol 2020; 42:431-450. [PMID: 32785751 DOI: 10.1007/s00281-020-00808-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022]
Abstract
Spontaneous preterm birth (PTB) and preterm pre-labor rupture of the membranes (pPROM) are major pregnancy complications. Although PTB and pPROM have common etiologies, they arise from distinct pathophysiologic pathways. Inflammation is a common underlying mechanism in both conditions. Balanced inflammation is required for fetoplacental growth; however, overwhelming inflammation (physiologic at term and pathologic at preterm) can lead to term and preterm parturition. A lack of effective strategies to control inflammation and reduce the risk of PTB and pPROM suggests that there are several modes of the generation of inflammation which may be dependent on the type of uterine tissue. The avascular fetal membrane (amniochorion), which provides structure, support, and protection to the intrauterine cavity, is one of the key contributors of inflammation. Localized membrane inflammation helps tissue remodeling during pregnancy. Two unique mechanisms that generate balanced inflammation are the progressive development of senescence (aging) and cyclic cellular transitions: epithelial to mesenchymal (EMT) and mesenchymal to epithelial (MET). The intrauterine build-up of oxidative stress at term or in response to risk factors (preterm) can accelerate senescence and promote a terminal state of EMT, resulting in the accumulation of inflammation. Inflammation degrades the matrix and destabilizes membrane function. Inflammatory mediators from damaged membranes are propagated via extracellular vesicles (EV) to maternal uterine tissues and transition quiescent maternal uterine tissues into an active state of labor. Membrane inflammation and its propagation are fetal signals that may promote parturition. This review summarizes the mechanisms of fetal membrane cellular senescence, transitions, and the generation of inflammation that contributes to term and preterm parturitions.
Collapse
Affiliation(s)
- Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA.
| | - Faranak Behnia
- Department of Obstetrics, Gynecology, and Reproductive Sciences, McGovern Medical School at the University of Texas Health Science Center at Houston, UT Health, Houston, Texas, USA
| | - Jossimara Polettini
- Universidade Federal da Fronteira Sul, Campus Passo Fundo, Rua Capitão Araujo, 20, Centro, Passo Fundo, Rio Grande do Sul, Brazil
| | - Lauren S Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, MRB 11.138, 301 301 University Blvd, Galveston, TX, 77555-1062, USA
| |
Collapse
|
16
|
Richardson L, Kim S, Menon R, Han A. Organ-On-Chip Technology: The Future of Feto-Maternal Interface Research? Front Physiol 2020; 11:715. [PMID: 32695021 PMCID: PMC7338764 DOI: 10.3389/fphys.2020.00715] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/29/2020] [Indexed: 12/17/2022] Open
Abstract
The placenta and fetal membrane act as a protective barrier throughout pregnancy while maintaining communication and nutrient exchange between the baby and the mother. Disruption of this barrier leads to various pregnancy complications, including preterm birth, which can have lasting negative consequences. Thus, understanding the role of the feto-maternal interface during pregnancy and parturition is vital to advancing basic and clinical research in the field of obstetrics. However, human subject studies are inherently difficult, and appropriate animal models are lacking. Due to these challenges, in vitro cell culture-based studies are most commonly utilized. However, the structure and functions of conventionally used in vitro 2D and 3D models are vastly different from the in vivo environment, making it difficult to fully understand the various factors affecting pregnancy as well as pathways and mechanisms contributing to term and preterm births. This limitation also makes it difficult to develop new therapeutics. The emergence of in vivo-like in vitro models such as organ-on-chip (OOC) platforms can better recapitulate in vivo functions and responses and has the potential to move this field forward significantly. OOC technology brings together two distinct fields, microfluidic engineering and cell/tissue biology, through which diverse human organ structures and functionalities can be built into a laboratory model that better mimics functions and responses of in vivo tissues and organs. In this review, we first provide an overview of the OOC technology, highlight two major designs commonly used in achieving multi-layer co-cultivation of cells, and introduce recently developed OOC models of the feto-maternal interface. As a vital component of this review, we aim to outline progress on the practicality and effectiveness of feto-maternal interface OOC (FM-OOC) models currently used and the advances they have fostered in obstetrics research. Lastly, we provide a perspective on the future basic research and clinical applications of FM-OOC models, and even those that integrate multiple organ systems into a single OOC system that may recreate intrauterine architecture in its entirety, which will accelerate our understanding of feto-maternal communication, induction of preterm labor, drug or toxicant permeability at this vital interface, and development of new therapeutic strategies.
Collapse
Affiliation(s)
- Lauren Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States.,Department of Electrical and Computer Engineering, College of Engineering, Texas A&M University, College Station, TX, United States.,Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
| | - Sungjin Kim
- Department of Electrical and Computer Engineering, College of Engineering, Texas A&M University, College Station, TX, United States.,Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Arum Han
- Department of Electrical and Computer Engineering, College of Engineering, Texas A&M University, College Station, TX, United States.,Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, TX, United States
| |
Collapse
|
17
|
Padron JG, Saito Reis CA, Kendal-Wright CE. The Role of Danger Associated Molecular Patterns in Human Fetal Membrane Weakening. Front Physiol 2020; 11:602. [PMID: 32625109 PMCID: PMC7311766 DOI: 10.3389/fphys.2020.00602] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 05/14/2020] [Indexed: 12/14/2022] Open
Abstract
The idea that cellular stress (including that precipitated by stretch), plays a significant role in the mechanisms initiating parturition, has gained considerable traction over the last decade. One key consequence of this cellular stress is the increased production of Danger Associated Molecular Patterns (DAMPs). This diverse family of molecules are known to initiate inflammation through their interaction with Pattern Recognition Receptors (PRRs) including, Toll-like receptors (TLRs). TLRs are the key innate immune system surveillance receptors that detect Pathogen Associated Molecular Patterns (PAMPs) during bacterial and viral infection. This is also seen during Chorioamnionitis. The activation of TLR commonly results in the activation of the pro-inflammatory transcription factor Nuclear Factor Kappa-B (NF-kB) and the downstream production of pro-inflammatory cytokines. It is thought that in the human fetal membranes both DAMPs and PAMPs are able, perhaps via their interaction with PRRs and the induction of their downstream inflammatory cascades, to lead to both tissue remodeling and weakening. Due to the high incidence of infection-driven Pre-Term Birth (PTB), including those that have preterm Premature Rupture of the Membranes (pPROM), the role of TLR in fetal membranes with Chorioamnionitis has been the subject of considerable study. Most of the work in this field has focused on the effect of PAMPs on whole pieces of fetal membrane and the resultant inflammatory cascade. This is important to understand, in order to develop novel prevention, detection, and therapeutic approaches, which aim to reduce the high number of mothers suffering from infection driven PTB, including those with pPROM. Studying the role of sterile inflammation driven by these endogenous ligands (DAMPs) activating PRRs system in the mesenchymal and epithelial cells in the amnion is important. These cells are key for the maintenance of the integrity and strength of the human fetal membranes. This review aims to (1) summarize the knowledge to date pertinent to the role of DAMPs and PRRs in fetal membrane weakening and (2) discuss the clinical potential brought by a better understanding of these pathways by pathway manipulation strategies.
Collapse
Affiliation(s)
- Justin G Padron
- Anatomy, Biochemistry and Physiology, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, United States
| | - Chelsea A Saito Reis
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States
| | - Claire E Kendal-Wright
- Natural Science and Mathematics, Chaminade University of Honolulu, Honolulu, HI, United States.,Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, United States
| |
Collapse
|
18
|
Richardson LS, Menon PR, Menon R. The effects of extracellular matrix rigidity on 3-dimensional cultures of amnion membrane cells. Placenta 2019; 90:82-89. [PMID: 32056556 DOI: 10.1016/j.placenta.2019.12.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/04/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023]
Abstract
INTRODUCTION To determine 3D growth of amnion membrane cells using soft substrate plates of various rigidities. METHODS Amnion epithelial (AEC) and mesenchymal cells (AMC) were cultured on 6-well soft substrate plates coated with matrigel and elastomer with rigidities of 0.5, 2, 8, 16, and 64 kPa (n = 3 each). Controls were cells in standard culture conditions. Cell morphology, spheroids' and sheets' formations and viability (bright field microscopy and crystal violet staining), and cellular transitions (vimentin/cytokeratin-18 [CK-18] ratios) were analyzed. A Student t-test was used for statistical analyses. RESULTS AECs in substrate rigidities between 2 and 8 kPa formed 3D features (spheroids and sheets) while retaining viability. Two kPa produced spheroids with epithelial characteristics (decrease in vimentin), and 8 kPa favored sheets. Transplantation and culture of AEC sheets with no matrix or elastomers, retained AECs' viability and maintained their epithelial characteristics. Optimum AMC growth was also between 2 and 8 kP A, with predominance of vimentin; however, AMCs did not form 3D structures. Lower and higher rigidities transitioned AMCs into AECs (decrease in vimentin). DISCUSSION Matrix rigidities between 2 and 8 kPa produced 3D structures of AECs (spheroids and sheets), resembling amnion membranes' morphology and exhibiting regenerative capacity in utero. Although AMCs grew in similar rigidities, a lack of 3D structures support their dispersed character in the membrane matrix. Extreme rigidities transitioned AMCs into AECs, suggesting that AMCs are transient cells (reservoirs) in the matrix required for remodeling. Compromises in matrix rigidity can cause membrane dysfunction and lead to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Lauren S Richardson
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA; Department of Neuroscience, Cell Biology & Anatomy, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| | - Poorna R Menon
- Clear Falls High School, 4380 Village Way, League City, TX, 77573, USA
| | - Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine & Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
19
|
Initiation of human parturition: signaling from senescent fetal tissues via extracellular vesicle mediated paracrine mechanism. Obstet Gynecol Sci 2019; 62:199-211. [PMID: 31338337 PMCID: PMC6629986 DOI: 10.5468/ogs.2019.62.4.199] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/06/2019] [Accepted: 03/20/2019] [Indexed: 12/19/2022] Open
Abstract
A better understanding of the underlying mechanisms by which signals from the fetus initiate human parturition is required. Our recent findings support the core hypothesis that oxidative stress (OS) and cellular senescence of the fetal membranes (amnion and chorion) trigger human parturition. Fetal membrane cell senescence at term is a natural physiological response to OS that occurs as a result of increased metabolic demands by the maturing fetus. Fetal membrane senescence is affected by the activation of the p38 mitogen activated kinase-mediated pathway. Similarly, various risk factors of preterm labor and premature rupture of the membranes also cause OS-induced senescence. Data suggest that fetal cell senescence causes inflammatory senescence-associated secretory phenotype (SASP) release. Besides SASP, high mobility group box 1 and cell-free fetal telomere fragments translocate from the nucleus to the cytosol in senescent cells, where they represent damage-associated molecular pattern markers (DAMPs). In fetal membranes, both SASPs and DAMPs augment fetal cell senescence and an associated ‘sterile’ inflammatory reaction. In senescent cells, DAMPs are encapsulated in extracellular vesicles, specifically exosomes, which are 30–150 nm particles, and propagated to distant sites. Exosomes traffic from the fetus to the maternal side and cause labor-associated inflammatory changes in maternal uterine tissues. Thus, fetal membrane senescence and the inflammation generated from this process functions as a paracrine signaling system during parturition. A better understanding of the premature activation of these signals can provide insights into the mechanisms by which fetal signals initiate preterm parturition.
Collapse
|
20
|
Barrett DW, John RK, Thrasivoulou C, Mata A, Deprest JA, Becker DL, David AL, Chowdhury TT. Targeting mechanotransduction mechanisms and tissue weakening signals in the human amniotic membrane. Sci Rep 2019; 9:6718. [PMID: 31040291 PMCID: PMC6491562 DOI: 10.1038/s41598-019-42379-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 03/29/2019] [Indexed: 11/24/2022] Open
Abstract
Mechanical and inflammatory signals in the fetal membrane play an important role in extracellular matrix (ECM) remodelling in order to dictate the timing of birth. We developed a mechanical model that mimics repetitive stretching of the amniotic membrane (AM) isolated from regions over the placenta (PAM) or cervix (CAM) and examined the effect of cyclic tensile strain (CTS) on mediators involved in mechanotransduction (Cx43, AKT), tissue remodelling (GAGs, elastin, collagen) and inflammation (PGE2, MMPs). In CAM and PAM specimens, the application of CTS increased GAG synthesis, PGE2 release and MMP activity, with concomitant reduction in collagen and elastin content. Co-stimulation with CTS and pharmacological agents that inhibit either Cx43 or AKT, differentially influenced collagen, GAG and elastin in a tissue-dependent manner. SHG confocal imaging of collagen fibres revealed a reduction in SHG intensity after CTS, with regions of disorganisation dependent on tissue location. CTS increased Cx43 and AKT protein and gene expression and the response could be reversed with either CTS, the Cx43 antisense or AKT inhibitor. We demonstrate that targeting Cx43 and AKT prevents strain-induced ECM damage and promotes tissue remodelling mechanisms in the AM. We speculate that a combination of inflammatory and mechanical factors could perturb typical mechanotransduction processes mediated by Cx43 signalling. Cx43 could therefore be a potential therapeutic target to prevent inflammation and preterm premature rupture of the fetal membranes.
Collapse
Affiliation(s)
- David W Barrett
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Rebecca K John
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Christopher Thrasivoulou
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Alvaro Mata
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK
| | - Jan A Deprest
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Anna L David
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Tina T Chowdhury
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, E1 4NS, UK.
| |
Collapse
|
21
|
Lim R, Barker G, Lappas M. Pellino 1 is a novel regulator of TNF and TLR signalling in human myometrial and amnion cells. J Reprod Immunol 2018; 127:24-35. [PMID: 29751216 DOI: 10.1016/j.jri.2018.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 02/15/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023]
Abstract
Preterm birth is the primary cause of neonatal deaths and morbidities. Pathological processes causally linked to preterm birth are inflammation and infection. Pellino-1 (Peli1) has previously been found to regulate the inflammatory response in non-gestational tissues in response to toll-like receptor (TLR) ligands and pro-inflammatory cytokines. The aims of this study were to determine the effect of labor on Peli1 expression in myometrium and fetal membranes, and the effect of Peli1 silencing by siRNA (siPELI1) on the production of pro-inflammatory and pro-labor mediators. The expression of Peli1 was found to be higher in myometrium and fetal membranes with term labor, compared to non-laboring samples. Peli1 mRNA and protein expression was also higher in amnion from women with preterm histological chorioamnionitis. In human primary myometrial cells, siPELI1 transfected cells showed a decrease in pro-inflammatory cytokine IL6, chemokines (CXCL8, CCL2) and adhesion molecule ICAM1 when in the presence of pro-inflammatory cytokine TNF, TLR2/6 ligand fsl-1, TLR5 ligand flagellin, and TLR3 ligand poly(I:C). Similarly in primary amnion cells, siPELI1 transfected cells decreased IL1B-induced expression and secretion of IL6 and CXCL8. In siPELI1 transfected myometrial cells, there was a decrease in prostaglandin PGF2α and its receptor, PTGFR mRNA expression when treated with TNF. There was a decrease in NF-κB RELA transcriptional activity in siPELI1 transfected cells in the presence of TNF, fsl-1 and flagellin, but not poly(I:C). Our study suggests a novel role for Peli1 in regulating pro-inflammatory and pro-labor mediators through TNF and TLR signalling.
Collapse
Affiliation(s)
- Ratana Lim
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Gillian Barker
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia; Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia.
| |
Collapse
|
22
|
Abstract
To generate new hypotheses, sometimes a "systems" approach is needed. In this review, I focus on the mitogen-activated kinase p38 because it has been recently shown to play an important role in the developmental programing and senescence of normal and stressed reproductive tissues. What follows is an overview of (i) pathways of p38 activation and their involvement in basic biological processes, (ii) evidence that p38 is involved in the homeostasis of reproductive tissues, (iii) how focus on p38 can be incorporated into investigation of normal and stressed pregnancies. Existence of excellent reviews will be mentioned as well as relevant animal models.
Collapse
Affiliation(s)
- Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Vermont, Burlington, VT, USA
| |
Collapse
|
23
|
Barrett DW, David AL, Thrasivoulou C, Mata A, Becker DL, Engels AC, Deprest JA, Chowdhury TT. Connexin 43 is overexpressed in human fetal membrane defects after fetoscopic surgery. Prenat Diagn 2016; 36:942-952. [PMID: 27568096 PMCID: PMC5082503 DOI: 10.1002/pd.4917] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/03/2016] [Accepted: 08/20/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE We examined whether surgically induced membrane defects elevate connexin 43 (Cx43) expression in the wound edge of the amniotic membrane (AM) and drives structural changes in collagen that affects healing after fetoscopic surgery. METHOD Cell morphology and collagen microstructure was investigated by scanning electron microscopy and second harmonic generation in fetal membranes taken from women who underwent fetal surgery. Immunofluoresence and real-time quantitative polymerase chain reaction was used to examine Cx43 expression in control and wound edge AM. RESULTS Scanning electron microscopy showed dense, helical patterns of collagen fibrils in the wound edge of the fetal membrane. This arrangement changed in the fibroblast layer with evidence of collagen fibrils that were highly polarised along the wound edge but not in control membranes. Cx43 was increased by 112.9% in wound edge AM compared with controls (p < 0.001), with preferential distribution in the fibroblast layer compared with the epithelial layer (p < 0.01). In wound edge AM, mesenchymal cells had a flattened morphology, and there was evidence of poor epithelial migration across the defect. Cx43 and COX-2 expression was significantly increased in wound edge AM compared with controls (p < 0.001). CONCLUSION Overexpression of Cx43 in the AM after fetal surgery induces morphological and structural changes in the collagenous matrix that may interfere with normal healing mechanisms. © 2016 The Authors. Prenatal Diagnosis published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- David W Barrett
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Anna L David
- Institute for Women's Health, University College London, London, UK
| | | | - Alvaro Mata
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - David L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Alex C Engels
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Jan A Deprest
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, Leuven, Belgium
| | - Tina T Chowdhury
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, UK.
| |
Collapse
|
24
|
Menon R, Bonney EA, Condon J, Mesiano S, Taylor RN. Novel concepts on pregnancy clocks and alarms: redundancy and synergy in human parturition. Hum Reprod Update 2016; 22:535-60. [PMID: 27363410 DOI: 10.1093/humupd/dmw022] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/16/2016] [Indexed: 12/19/2022] Open
Abstract
The signals and mechanisms that synchronize the timing of human parturition remain a mystery and a better understanding of these processes is essential to avert adverse pregnancy outcomes. Although our insights into human labor initiation have been informed by studies in animal models, the timing of parturition relative to fetal maturation varies among viviparous species, indicative of phylogenetically different clocks and alarms; but what is clear is that important common pathways must converge to control the birth process. For example, in all species, parturition involves the transition of the myometrium from a relaxed to a highly excitable state, where the muscle rhythmically and forcefully contracts, softening the cervical extracellular matrix to allow distensibility and dilatation and thus a shearing of the fetal membranes to facilitate their rupture. We review a number of theories promulgated to explain how a variety of different timing mechanisms, including fetal membrane cell senescence, circadian endocrine clocks, and inflammatory and mechanical factors, are coordinated as initiators and effectors of parturition. Many of these factors have been independently described with a focus on specific tissue compartments.In this review, we put forth the core hypothesis that fetal membrane (amnion and chorion) senescence is the initiator of a coordinated, redundant signal cascade leading to parturition. Whether modified by oxidative stress or other factors, this process constitutes a counting device, i.e. a clock, that measures maturation of the fetal organ systems and the production of hormones and other soluble mediators (including alarmins) and that promotes inflammation and orchestrates an immune cascade to propagate signals across different uterine compartments. This mechanism in turn sensitizes decidual responsiveness and eventually promotes functional progesterone withdrawal in the myometrium, leading to increased myometrial cell contraction and the triggering of parturition. Linkage of these processes allows convergence and integration of the gestational clocks and alarms, prompting a timely and safe birth. In summary, we provide a comprehensive synthesis of the mediators that contribute to the timing of human labor. Integrating these concepts will provide a better understanding of human parturition and ultimately improve pregnancy outcomes.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Perinatal Research, The University of Texas Medical Branch at Galveston, 301 University Blvd., MRB, Room 11.138, Galveston, TX 77555-1062, USA
| | - Elizabeth A Bonney
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont College of Medicine, 792 College Parkway, Fanny Allen Campus, Suite 101, Colchester, Burlington, VT 05446, USA
| | - Jennifer Condon
- Department of Obstetrics and Gynecology, Wayne State University, Perinatal Research Branch, NICHD, Detroit, MI 48201, USA
| | - Sam Mesiano
- Department of Reproductive Biology and Obstetrics and Gynecology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH 44106, USA
| | - Robert N Taylor
- Department of Obstetrics and Gynecology, Medical Center Boulevard, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
25
|
Balaji S, Watson CL, Ranjan R, King A, Bollyky PL, Keswani SG. Chemokine Involvement in Fetal and Adult Wound Healing. Adv Wound Care (New Rochelle) 2015; 4:660-672. [PMID: 26543680 DOI: 10.1089/wound.2014.0564] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Significance: Fetal wounds heal with a regenerative phenotype that is indistinguishable from surrounding skin with restored skin integrity. Compared to this benchmark, all postnatal wound healing is impaired and characterized by scar formation. The biologic basis of the fetal regenerative phenotype can serve as a roadmap to recapitulating regenerative repair in adult wounds. Reduced leukocyte infiltration, likely mediated, in part, through changes in the chemokine milieu, is a fundamental feature of fetal wound healing. Recent Advances: The contributions of chemokines to wound healing are a topic of active investigation. Recent discoveries have opened the possibility of targeting chemokines therapeutically to treat disease processes and improve healing capability, including the possibility of achieving a scarless phenotype in postnatal wounds. Critical Issues: Successful wound healing is a complex process, in which there is a significant interplay between multiple cell types, signaling molecules, growth factors, and extracellular matrix. Chemokines play a crucial role in this interplay and have been shown to have different effects in various stages of the healing process. Understanding how these chemokines are locally produced and regulated during wound healing and how the chemokine milieu differs in fetal versus postnatal wounds may help us identify ways in which we can target chemokine pathways. Future Directions: Further studies on the role of chemokines and their role in the healing process will greatly advance the potential for using these molecules as therapeutic targets.
Collapse
Affiliation(s)
- Swathi Balaji
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Carey L. Watson
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Rajeev Ranjan
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Alice King
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio
| | - Paul L. Bollyky
- Division of Infectious Diseases, Department of Medicine, Stanford School of Medicine, Palo Alto, California
| | - Sundeep G. Keswani
- Laboratory for Regenerative Wound Healing, Division of Pediatric, General, Thoracic and Fetal Surgery, Cincinnati Children's Hospital and the University of Cincinnati, College of Medicine, Cincinnati, Ohio
| |
Collapse
|
26
|
Pavlová T, Novák J, Bienertová-Vašků J. The role of visfatin (PBEF/Nampt) in pregnancy complications. J Reprod Immunol 2015; 112:102-10. [PMID: 26451650 DOI: 10.1016/j.jri.2015.09.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 07/13/2015] [Accepted: 09/17/2015] [Indexed: 01/05/2023]
Abstract
Visfatin (PBEF/Nampt) is an adipocytokine that exerts pleiotropic effects within the human body, particularly affecting its metabolism and immunity. Visfatin was originally identified as being secreted by peripheral blood lymphocytes acting as a pre-B-cell colony-enhancing factor (PBEF). However, it was subsequently reported to be expressed in almost every tissue of the human body, with visceral fat deposits being the main source of visfatin. In addition to its secreted form, visfatin may also be found intracellularly where it functions as a nicotinamide phosphoribosyltransferase (Nampt). Visfatin maternal plasma concentrations increase during pregnancy, suggesting its important role in this complicated process. Alterations in visfatin level also take place in patients during pregnancy complications. This review focuses on the ones that most commonly occur in connection with visfatin: preterm labor, pre-eclampsia and gestational diabetes mellitus. The review aims to provide a better understanding of the role of visfatin during pregnancy and the causes of its alteration in maternal plasma, highlighting the potential use of visfatin as a diagnostic marker of pregnancy complications in the future.
Collapse
Affiliation(s)
- Tereza Pavlová
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A18, Brno 62500, Czech Republic.
| | - Jan Novák
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A18, Brno 62500, Czech Republic.
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Building A18, Brno 62500, Czech Republic; Regional Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Žlutý kopec 7, Brno 65653, Czech Republic.
| |
Collapse
|
27
|
Heng YJ, Liong S, Permezel M, Rice GE, Di Quinzio MKW, Georgiou HM. Human cervicovaginal fluid biomarkers to predict term and preterm labor. Front Physiol 2015; 6:151. [PMID: 26029118 PMCID: PMC4429550 DOI: 10.3389/fphys.2015.00151] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 04/27/2015] [Indexed: 01/06/2023] Open
Abstract
Preterm birth (PTB; birth before 37 completed weeks of gestation) remains the major cause of neonatal morbidity and mortality. The current generation of biomarkers predictive of PTB have limited utility. In pregnancy, the human cervicovaginal fluid (CVF) proteome is a reflection of the local biochemical milieu and is influenced by the physical changes occurring in the vagina, cervix and adjacent overlying fetal membranes. Term and preterm labor (PTL) share common pathways of cervical ripening, myometrial activation and fetal membranes rupture leading to birth. We therefore hypothesize that CVF biomarkers predictive of labor may be similar in both the term and preterm labor setting. In this review, we summarize some of the existing published literature as well as our team's breadth of work utilizing the CVF for the discovery and validation of putative CVF biomarkers predictive of human labor. Our team established an efficient method for collecting serial CVF samples for optimal 2-dimensional gel electrophoresis resolution and analysis. We first embarked on CVF biomarker discovery for the prediction of spontaneous onset of term labor using 2D-electrophoresis and solution array multiple analyte profiling. 2D-electrophoretic analyses were subsequently performed on CVF samples associated with PTB. Several proteins have been successfully validated and demonstrate that these biomarkers are associated with term and PTL and may be predictive of both term and PTL. In addition, the measurement of these putative biomarkers was found to be robust to the influences of vaginal microflora and/or semen. The future development of a multiple biomarker bed-side test would help improve the prediction of PTB and the clinical management of patients.
Collapse
Affiliation(s)
- Yujing J Heng
- Department of Pathology, Harvard Medical School and Beth Israel Deaconess Medical Center Boston, MA, USA
| | - Stella Liong
- Department of Obstetrics and Gynaecology, University of Melbourne Melbourne, VIC, Australia ; Mercy Perinatal Research Centre, Mercy Hospital for Women Heidelberg, VIC, Australia
| | - Michael Permezel
- Department of Obstetrics and Gynaecology, University of Melbourne Melbourne, VIC, Australia ; Mercy Perinatal Research Centre, Mercy Hospital for Women Heidelberg, VIC, Australia
| | - Gregory E Rice
- University of Queensland Centre for Clinical Research Herston, QLD, Australia
| | - Megan K W Di Quinzio
- Department of Obstetrics and Gynaecology, University of Melbourne Melbourne, VIC, Australia ; Mercy Perinatal Research Centre, Mercy Hospital for Women Heidelberg, VIC, Australia
| | - Harry M Georgiou
- Department of Obstetrics and Gynaecology, University of Melbourne Melbourne, VIC, Australia ; Mercy Perinatal Research Centre, Mercy Hospital for Women Heidelberg, VIC, Australia
| |
Collapse
|
28
|
Chowdhury B, David AL, Thrasivoulou C, Becker DL, Bader DL, Chowdhury TT. Tensile strain increased COX-2 expression and PGE2 release leading to weakening of the human amniotic membrane. Placenta 2014; 35:1057-64. [PMID: 25280972 DOI: 10.1016/j.placenta.2014.09.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 07/14/2014] [Accepted: 09/11/2014] [Indexed: 11/25/2022]
Abstract
INTRODUCTION There is evidence that premature rupture of the fetal membrane at term/preterm is a result of stretch and tissue weakening due to enhanced prostaglandin E2 (PGE2) production. However, the effect of tensile strain on inflammatory mediators and the stretch sensitive protein connexin-43 (Cx43) has not been examined. We determined whether the inflammatory environment influenced tissue composition and response of the tissue to tensile strain. METHODS Human amniotic membranes isolated from the cervix (CAM) or placenta regions (PAM) were examined by second harmonic generation to identify collagen orientation and subjected to tensile testing to failure. In separate experiments, specimens were subjected to cyclic tensile strain (2%, 1 Hz) for 24 h. Specimens were examined for Cx43 by immunofluorescence confocal microscopy and expression of COX-2 and Cx43 by RT-qPCR. PGE2, collagen, elastin and glycosaminoglycan (GAG) levels were analysed by biochemical assay. RESULTS Values for tensile strength were significantly higher in PAM than CAM with mechanical parameters dependent on collagen orientation. Gene expression for Cx43 and COX-2 was enhanced by tensile strain leading to increased PGE2 release and GAG levels in PAM and CAM when compared to unstrained controls. In contrast, collagen and elastin content was reduced by tensile strain in PAM and CAM. DISCUSSION Fibre orientation has a significant effect on amniotic strength. Tensile strain increased Cx43/COX-2 expression and PGE2 release resulting in tissue softening mediated by enhanced GAG levels and a reduction in collagen/elastin content. CONCLUSION A combination of inflammatory and mechanical factors may disrupt amniotic membrane biomechanics and matrix composition.
Collapse
Affiliation(s)
- B Chowdhury
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK
| | - A L David
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK
| | - C Thrasivoulou
- Department of Cell and Developmental Biology, UCL, Gower Street, London WC1E 6BT, UK
| | - D L Becker
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11, Mandalay Road, Singapore
| | - D L Bader
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; Faculty of Health Sciences, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK
| | - T T Chowdhury
- Institute of Bioengineering, School of Engineering and Material Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK.
| |
Collapse
|
29
|
Lee YH, Shynlova O, Lye SJ. Stretch-induced human myometrial cytokines enhance immune cell recruitment via endothelial activation. Cell Mol Immunol 2014; 12:231-42. [PMID: 24882387 DOI: 10.1038/cmi.2014.39] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 04/16/2014] [Accepted: 05/05/2014] [Indexed: 01/05/2023] Open
Abstract
Spontaneous term labour is associated with amplified inflammatory events in the myometrium including cytokine production and leukocyte infiltration; however, potential mechanisms regulating such events are not fully understood. We hypothesized that mechanical stretch of the uterine wall by the growing fetus facilitates peripheral leukocyte extravasation into the term myometrium through the release of various cytokines by uterine myocytes. Human myometrial cells (hTERT-HM) were subjected to static mechanical stretch; stretch-conditioned media was collected and analysed using 48-plex Luminex assay and ELISA. Effect of stretch-conditioned media on cell adhesion molecule expression of human uterine microvascular endothelial cells (UtMVEC-Myo) was detected by quantitative polymerase chain reaction (qPCR) and flow cytometry; functional assays testing leukocyte-endothelial interactions: adhesion of leukocytes to endothelial cells and transendothelial migration of calcein-labelled primary human neutrophils as well as migration of THP-1 monocytic cells were assessed by fluorometry. The current in vitro study demonstrated that mechanical stretch (i) directly induces secretion of multiple cytokines and chemokines by hTERT-HM cells (IL-6, CXCL8, CXCL1, migration inhibitory factor (MIF), VEGF, G-CSF, IL-12p70, bFGF and platelet-derived growth factor subunit B (PDGF-bb), P<0.05); stretch-induced cytokines (ii) enhance leukocyte adhesion to the endothelium of the surrounding uterine microvasculature by (iii) inducing the expression of endothelial cell adhesion molecules and (iv) directing the transendothelial migration of peripheral leukocytes. (vi) Chemokine-neutralizing antibodies and broad-spectrum chemokine inhibitor block leukocyte migration. Our data provide a proof of mechanical regulation for leukocyte recruitment from the uterine blood vessels to the myometrium, suggesting a putative mechanism for the leukocyte infiltrate into the uterus during labour and postpartum involution.
Collapse
Affiliation(s)
- Yu-Hui Lee
- 1] Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada [2] Department of Physiology, University of Toronto, Toronto, Canada
| | - Oksana Shynlova
- 1] Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada [2] Department of Obstetrics & Gynecology, University of Toronto, Toronto, Canada
| | - Stephen J Lye
- 1] Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada [2] Department of Physiology, University of Toronto, Toronto, Canada [3] Department of Obstetrics & Gynecology, University of Toronto, Toronto, Canada
| |
Collapse
|
30
|
Blanchon L, Accoceberry M, Belville C, Delabaere A, Prat C, Lemery D, Sapin V, Gallot D. [Rupture of membranes: pathophysiology, diagnosis, consequences and management]. ACTA ACUST UNITED AC 2013; 42:105-16. [PMID: 23395133 DOI: 10.1016/j.jgyn.2012.12.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 12/22/2012] [Accepted: 12/28/2012] [Indexed: 10/27/2022]
Abstract
Rupture of membranes (ROM) depends on mechanical stretch, extracellular matrix components imbalance and increased apoptosis. It occurs in 2 to 3% of all pregnancies before 37 weeks' gestation (WG) and in up to 10% at term. Main consequences are labor induction and risk of maternal-fetal infection. ROM is associated with one third of preterm births and about 20% of perinatal mortality. This review deals with recent knowledge concerning ROM including diagnosis and management. In many cases, ROM is easily identified by clinical examination. In other cases, the use of vaginal pH appears to be less efficient than the use of immunochromatographic strips based on IGFBP-1 or PAMG-1 detection. Before 34WG, conservative management consists in in utero transfer, antibioprophylaxis and corticosteroids. After 37WG, delivery is the most appropriate option. Between 34 and 37WG, recent studies demonstrate that induction of labour does not improve pregnancy outcomes. Therefore, expectant management can be the first option between 34 and 37WG when no active infection is suspected especially in case of unfavourable cervix.
Collapse
Affiliation(s)
- L Blanchon
- R2D2-EA7281, faculté de médecine, université d'Auvergne, place Henri-Dunant, 63000 Clermont-Ferrand, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kim HR, Han RX, Diao YF, Park CS, Jin DI. Epigenetic characterization of the PBEF and TIMP-2 genes in the developing placentae of normal mice. BMB Rep 2011; 44:535-40. [PMID: 21871178 DOI: 10.5483/bmbrep.2011.44.8.535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reprogramming errors, which appear frequently in cloned animals, are reflected by aberrant gene expression. We previously reported the aberrant expression of TIMP-2 and PBEF in cloned placenta and differential expression of PBEF genes during pregnancy. To examine the epigenetic modifications that regulate dynamic gene expression in developing placentae, we herein analyzed the mRNA and protein expression levels of PBEF and TIMP-2 in the placentae of normal mice during pregnancy and then examined potential correlations with epigenetic modifications. DNA methylation pattern analysis revealed no difference, but ChIP assays using antibodies against H3-K9/K14 and H4-K5 histone acetylation revealed that the H3-K9/K14 acetylation levels, but not the H4-K5 acetylation levels, of the TIMP-2 and PBEF loci were significantly correlated with their gene expression levels during placentation in normal mice. These results suggest that epigenetic changes may regulate gene expression level in the developing placentae of normal mice and that inappropriate epigenetic reprogramming might be one cause of the abnormal placentae seen in cloned animals.
Collapse
Affiliation(s)
- Hong-Rye Kim
- Department of Animal Science & Biotechnology, Research Center for Transgenic Cloned Pig, Chungnam National University, Deajeon, Korea
| | | | | | | | | |
Collapse
|
32
|
McDonald C, Siatskas C, C.A. Bernard C. The emergence of amnion epithelial stem cells for the treatment of Multiple Sclerosis. Inflamm Regen 2011. [DOI: 10.2492/inflammregen.31.256] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
33
|
Rumsey JW, Das M, Bhalkikar A, Stancescu M, Hickman JJ. Tissue engineering the mechanosensory circuit of the stretch reflex arc: sensory neuron innervation of intrafusal muscle fibers. Biomaterials 2010; 31:8218-27. [PMID: 20708792 DOI: 10.1016/j.biomaterials.2010.07.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2010] [Accepted: 07/04/2010] [Indexed: 11/19/2022]
Abstract
The sensory circuit of the stretch reflex arc, composed of specialized intrafusal muscle fibers and type Ia proprioceptive sensory neurons, converts mechanical information regarding muscle length and stretch to electrical action potentials and relays them to the central nervous system. Utilizing a non-biological substrate, surface patterning photolithography and a serum-free medium formulation a co-culture system was developed that facilitated functional interactions between intrafusal muscle fibers and sensory neurons. The presence of annulospiral wrappings (ASWs) and flower-spray endings (FSEs), both physiologically relevant morphologies in sensory neuron-intrafusal fiber interactions, were demonstrated and quantified using immunocytochemistry. Furthermore, two proposed components of the mammalian mechanosensory transduction system, BNaC1 and PICK1, were both identified at the ASWs and FSEs. To verify functionality of the mechanoreceptor elements the system was integrated with a MEMS cantilever device, and Ca(2+) currents were imaged along the length of an axon innervating an intrafusal fiber when stretched by cantilever deflection. This system provides a platform for examining the role of this mechanosensory complex in the pathology of myotonic and muscular dystrophies, peripheral neuropathy, and spasticity inducing diseases like Parkinson's. These studies will also assist in engineering fine motor control for prosthetic devices by improving our understanding of mechanosensitive feedback.
Collapse
Affiliation(s)
- John W Rumsey
- NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | | | | | | | | |
Collapse
|
34
|
Mazaki-Tovi S, Romero R, Kim SK, Vaisbuch E, Kusanovic JP, Erez O, Chaiworapongsa T, Gotsch F, Mittal P, Nhan-Chang CL, Than NG, Gomez R, Nien JK, Edwin SS, Pacora P, Yeo L, Hassan SS. Could alterations in maternal plasma visfatin concentration participate in the phenotype definition of preeclampsia and SGA? J Matern Fetal Neonatal Med 2010; 23:857-68. [PMID: 19900033 PMCID: PMC3554253 DOI: 10.3109/14767050903301017] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE Women with preeclampsia and those who delivered a small-for-gestational-age (SGA) neonate share several mechanisms of disease, including chronic uteroplacental ischemia and failure of physiologic transformation of the spiral arteries. However, the clinical manifestation of these obstetrical syndromes is remarkably different. It has been proposed that an altered maternal metabolic state, as well as a unique circulating cytokines milieu, predispose women to develop either preeclampsia or SGA. Compelling evidence suggests that adipose tissue orchestrates both metabolic pathways and immunological responses via the production of adipokines. Visfatin is a novel adipocytokine with metabolic and immunomodulating properties. The objective of this study was to determine whether preeclampsia and SGA are associated with alterations in maternal circulating visfatin concentrations. METHODS This cross-sectional study included pregnant women in the following groups: (1) normal pregnancy (n = 158); (2) patients with preeclampsia (n = 43) of which 32 had an AGA and 11 had an SGA neonate; (3) patients without preeclampsia who delivered an SGA neonate (n = 55). Maternal plasma visfatin concentrations were measured by ELISA. Nonparametric tests and multiple linear regression analysis were used. RESULTS (1) Women who delivered an SGA neonate had a higher median maternal plasma visfatin concentration than those with a normal pregnancy (20.0 ng/ml, interquartile range: 17.2-24.6 vs. 15.2 ng/ml, 12.1-19.2, respectively; P < 0.001) and than those with preeclampsia (14.5 ng/ml, 12.5-18.7; P < 0.001); (2) the median maternal plasma visfatin concentration did not differ significantly between patients with preeclampsia and those with a normal pregnancy (P = 0.8); (3) among patients with preeclampsia, there was no significant difference in the median maternal plasma visfatin concentration between those with or without an SGA neonate (P = 0.5); (4) in a linear regression model, delivery of an SGA neonate and pregestational body mass index were independently associated with increased visfatin concentration after adjustment for confounding factors (maternal age, smoking, gestational age at blood collection and the presence of preeclampsia or SGA). CONCLUSION (1) Patients with SGA, but not those with preeclampsia, had a higher maternal plasma visfatin concentration than those with a normal pregnancy; (2) this finding suggests differential involvement of visfatin in SGA and preeclampsia; (3) we propose that changes in circulating maternal visfatin concentration may be implicated in the phenotypic definitions and distinction of preeclampsia and SGA.
Collapse
Affiliation(s)
- Shali Mazaki-Tovi
- Perinatology Research Branch, Intramural Division, NICHD/NIH/DHHS, Hutzel Women's Hospital, Bethesda, MD, and Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Kendal-Wright CE, Hubbard D, Gowin-Brown J, Bryant-Greenwood GD. Stretch and inflammation-induced Pre-B cell colony-enhancing factor (PBEF/Visfatin) and Interleukin-8 in amniotic epithelial cells. Placenta 2010; 31:665-74. [PMID: 20598369 DOI: 10.1016/j.placenta.2010.06.007] [Citation(s) in RCA: 18] [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: 03/16/2010] [Revised: 06/04/2010] [Accepted: 06/09/2010] [Indexed: 11/18/2022]
Abstract
Preterm birth continues to be a growing problem in the USA. Although approximately half of preterm births are caused by intrauterine infection, uterine over-distension is also a cause. In this study we have compared the effects of static stretch, cyclic stretch/release and an inflammatory stimulus alone and in combination on the expression of Pre-B cell colony-enhancing factor (PBEF) and IL-8 in primary amniotic epithelial cells (AEC). We then sought to identify some of the mechanism(s) by which these cells respond to stretching stimuli. We show that cyclic stretch/release is a more robust stimulus for both PBEF and IL-8 than static stretch. Cyclic stretch/release increased both intracellular and secreted PBEF and a combination of both types of stretch was a more robust stimulus to PBEF that IL-8. However, when an inflammatory stimulus (IL-1beta) was added to either kind of stretch, the effect on IL-8 was much greater than that on PBEF. Thus, different kinds of stretch affect the expression of these two cytokines from AEC, but inflammation is a much stronger stimulus of IL-8 than PBEF, agreeing with its primary role as a chemokine. Although the AEC showed morphological signs of increased cellular stress during stretching, blocking reactive oxygen species (ROS) had little effect. However, blocking integrin binding to fibronectin significantly reduced the responses of both PBEF and IL-8 to cyclic stretch/release. The increased PBEF, both intracellularly and secreted, suggests that it functions both to increase the metabolism of the cells, at the same time as stimulating further the cytokine cascade leading to parturition.
Collapse
Affiliation(s)
- C E Kendal-Wright
- Department of Obstetrics, Gynecology and Women's Health, John A. Burns School of Medicine, University of Hawai'i, Honolulu, HI 96813, USA.
| | | | | | | |
Collapse
|
36
|
Mazaki-Tovi S, Romero R, Vaisbuch E, Kim SK, Kusanovic JP, Chaiworapongsa T, Mittal P, Dong Z, Pacora P, Yeo L, Hassan SS. Evidence for differential regulation of the adipokine visfatin in the maternal and fetal compartments in normal spontaneous labor at term. J Perinat Med 2010; 38:281-8. [PMID: 20146661 PMCID: PMC3424273 DOI: 10.1515/jpm.2010.045] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Visfatin, a novel adipokine with metabolic and immunoregulatory properties, has been implicated in the regulation of fetal growth, as well as in preterm labor. A gap in knowledge is whether spontaneous labor at term is associated with changes in the maternal and fetal concentrations of visfatin. The aim of this study was to determine if the presence of labor at term is associated with alterations in maternal and neonatal plasma visfatin concentrations. STUDY DESIGN This cross-sectional study included 50 normal pregnant women at term and their appropriate-for-gestational age (AGA) neonates in the following groups: 1) 25 mother-neonate pairs delivered by elective cesarean section without spontaneous labor, and 2) 25 mother-neonate pairs who delivered vaginally following spontaneous labor. Maternal plasma and cord blood visfatin concentrations were determined by ELISA. Non-parametric statistics were used for analyses. RESULTS 1) The median visfatin concentration was higher in umbilical cord plasma of neonates born following a spontaneous labor at term than that of those who were born by an elective cesarean section (P=0.02); 2) in contrast, the median maternal plasma visfatin concentration did not differ significantly between patients with and without labor (P=0.44); and 3) there was a significant correlation between umbilical cord plasma concentration of visfatin and both maternal visfatin concentration (r=0.54, P=0.005) and gestational age (GA) at delivery (r=0.58; P=0.002) only in the absence of labor. CONCLUSION Term labor is associated with increased fetal, but not maternal, circulating visfatin concentrations. Previous reports indicate that preterm labor leading to preterm delivery is characterized by an increase in maternal plasma concentrations of visfatin. The observations reported herein support the view that there are fundamental differences in the endocrine and metabolic adaptations in normal labor at term and preterm labor.
Collapse
Affiliation(s)
- Shali Mazaki-Tovi
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Roberto Romero
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Edi Vaisbuch
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Sun Kwon Kim
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
| | - Juan Pedro Kusanovic
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Tinnakorn Chaiworapongsa
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Pooja Mittal
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Zhong Dong
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
| | - Percy Pacora
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
| | - Lami Yeo
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| | - Sonia S. Hassan
- Perinatology Research Branch, NICHD/NIH/DHHS, Bethesda, MD, and Detroit, MI
- Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI
| |
Collapse
|
37
|
Mazaki-Tovi S, Romero R, Vaisbuch E, Erez O, Chaiworapongsa T, Mittal P, Kim SK, Pacora P, Gotsch F, Dong Z, Hassan SS, Kusanovic JP. Maternal plasma visfatin in preterm labor. J Matern Fetal Neonatal Med 2009; 22:693-704. [PMID: 19572235 PMCID: PMC6656365 DOI: 10.1080/14767050902994788] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Visfatin, a novel adipokine with diabetogenic and immunoregulatory properties, has been implicated in the pathophysiology of insulin resistance, as well as in various acute and chronic inflammatory disorders. We have previously reported that amniotic fluid concentrations of visfatin are higher in patients with preterm labor (PTL) and intra-amniotic infection than in patients with PTL without infection. The aim of this study was to determine whether spontaneous PTL with intact membranes and intra-amniotic infection/inflammation (IAI) is associated with changes in maternal plasma circulating visfatin concentrations. STUDY DESIGN This cross-sectional study included patients in the following groups: (1) normal pregnant women (n = 123); (2) patients with an episode of PTL and intact membranes without IAI who delivered at term (n = 57); (3) PTL without IAI who delivered preterm (n = 47); and (4) PTL with IAI who delivered preterm (n = 57). Plasma visfatin concentrations were determined by ELISA. Non-parametric statistics were used for analysis. RESULTS (1) PTL with IAI leading to preterm delivery was associated with a higher median maternal plasma concentration of visfatin than normal pregnancy; (2) among patients with PTL, those with IAI had the highest median maternal concentration of visfatin; (3) the changes in maternal plasma visfatin remained significant after adjusting for maternal age, body mass index, gestational age at sampling, and birth weight. CONCLUSION (1) PTL with IAI is characterized by high maternal circulating visfatin concentrations; (2) these findings suggest that visfatin plays a role in the regulation of the metabolic adaptations to insults resulting in PTL in the context of IAI.
Collapse
Affiliation(s)
- Shali Mazaki-Tovi
- Intramural Division, Perinatology Research Branch, NICHD/NIH/DHHS, Hutzel Women's Hospital, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mazaki-Tovi S, Romero R, Kusanovic JP, Vaisbuch E, Erez O, Than NG, Chaiworapongsa T, Nhan-Chang CL, Pacora P, Gotsch F, Yeo L, Kim SK, Edwin SS, Hassan SS, Mittal P. Visfatin in human pregnancy: maternal gestational diabetes vis-à-vis neonatal birthweight. J Perinat Med 2009; 37:218-31. [PMID: 19099366 PMCID: PMC3504974 DOI: 10.1515/jpm.2009.053] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Adipose tissue dysfunction, characterized by dysregulation of adipokines production and/or secretion, has been implicated in the pathophysiology of type-2 diabetes mellitus, a metabolic complication closely related to gestational diabetes mellitus (GDM). Recently, an association between circulating maternal visfatin, a novel adipokine with metabolic and immunoregulatory properties, and impaired glucose metabolism as well as with altered fetal growth, has been proposed. The aims of this study were to determine whether there is an association between maternal plasma visfatin concentration, GDM, and a large-for-gestational-age (LGA) newborn. STUDY DESIGN This cross-sectional study, included pregnant women at term in the following groups: 1) normal pregnancy and an appropriate-for-gestational-age (AGA) neonate (n=54); 2) normal pregnancy and an LGA newborn (n=47); 3) GDM and an AGA newborn (n=56); 4) GDM and an LGA newborn (n=45). The study population was further stratified by first trimester BMI (<25 vs. > or =25 kg/m(2)). Maternal plasma visfatin concentration was determined by ELISA. Parametric and non-parametric statistics were used for analysis. RESULTS 1) Among women who delivered an AGA neonate, the median maternal plasma concentration of visfatin was higher in patients with GDM than in those with a normal pregnancy; 2) Among women with a normal pregnancy, those who delivered an LGA neonate had a higher median maternal plasma visfatin concentration than those who delivered an AGA neonate; 3) among patients with normal BMI, there were no significant differences in the median maternal plasma visfatin concentration between the four study groups; and 4) maternal GDM, as well as delivery of an LGA neonate were independently associated with a higher maternal plasma visfatin concentrations. CONCLUSION The linkage between increased maternal circulating visfatin and the presence of GDM or delivery of an LGA neonate supports the hypothesis that perturbation of adipokines homeostasis may play a role in the pathophysiology of GDM or excess fetal growth.
Collapse
Affiliation(s)
- Shali Mazaki-Tovi
- Perinatology Research Branch, Intramural Division, NICHD/NIH/DHHS, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|