1
|
Holme JA, Myhre O, Øvrevik J. Adverse neurodevelopment in children associated with prenatal exposure to fine particulate matter (PM 2.5) - Possible roles of polycyclic aromatic hydrocarbons (PAHs) and mechanisms involved. Reprod Toxicol 2024; 130:108718. [PMID: 39276806 DOI: 10.1016/j.reprotox.2024.108718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Prenatal exposure to ambient fine particles (PM2.5) and polycyclic aromatic hydrocarbons (PAHs) has been associated with adverse birth outcomes including neurodevelopmental effects with cognitive and/or behavioral implications in early childhood. As a background we first briefly summarize human studies on PM2.5 and PAHs associated with adverse birth outcomes and modified neurodevelopment. Next, we add more specific information from animal studies and in vitro studies and elucidate possible biological mechanisms. More specifically we focus on the potential role of PAHs attached to PM2.5 and explore whether effects of these compounds may arise from disturbance of placental function or more directly by interfering with neurodevelopmental processes in the fetal brain. Possible molecular initiating events (MIEs) include interactions with cellular receptors such as the aryl hydrocarbon receptor (AhR), beta-adrenergic receptors (βAR) and transient receptor potential (TRP)-channels resulting in altered gene expression. MIE linked to the binding of PAHs to cytochrome P450 (CYP) enzymes and formation of reactive electrophilic metabolites are likely less important. The experimental animal and in vitro studies support the epidemiological findings and suggest steps involved in mechanistic pathways explaining the associations. An overall evaluation of the doses/concentrations used in experimental studies combined with the mechanistic understanding further supports the hypothesis that prenatal PAHs exposure may cause adverse outcomes (AOs) linked to human neurodevelopment. Several MIEs will likely occur simultaneously in various cells/tissues involving several key events (KEs) which relative importance will depend on dose, time, tissue, genetics, other environmental factors, and neurodevelopmental endpoint in study.
Collapse
Affiliation(s)
- Jørn A Holme
- Department of Air quality and Noise, Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box PO Box 222 Skøyen, Oslo 0213, Norway.
| | - Oddvar Myhre
- Department of Chemical Toxicology, Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo 0213, Norway
| | - Johan Øvrevik
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, PO Box 1066 Blindern, Oslo 0316, Norway; Division of Climate and Environmental Health, Norwegian Institute of Public Health, PO Box 222 Skøyen, Oslo 0213, Norway
| |
Collapse
|
2
|
Bappoo N, Tongpob Y, Hakim M, Myers J, Panting E, Chapman KE, Thomson AJW, Moran CM, Kelsey LJ, Srinivasan V, James JL, Clark AR, Doyle BJ, Wyrwoll CS. Feto-placental vascular structure and in silico haemodynamics: Of mice, rats, and human. Placenta 2024; 158:175-184. [PMID: 39476476 DOI: 10.1016/j.placenta.2024.10.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/31/2024] [Accepted: 10/23/2024] [Indexed: 12/11/2024]
Abstract
INTRODUCTION The complex arborization of the feto-placental vasculature is crucial for optimal fetal nutrition, waste exchange and ultimately, development. Ethical and experimental limitations constrain research into the human placenta, hence experimental animal models such as mice and rats, are crucial to understand placental function. It is unclear how well the mouse and rat feto-placental vascular structure emulates human. Moreover, the implications of differences in vascular structure, especially in arborization, for placental function remain unclear. METHODS We use micro-computed tomography imaging, high frequency Doppler ultrasound and computational fluid dynamics to characterize feto-placental vasculature structure and haemodynamics in mice, rats, and human. RESULTS Our data suggest that despite structural differences between rat and mouse placenta, haemodynamics are similar and that both hold applicability to investigating feto-placental structure and function. We show that human cotyledons demonstrate vascularity-dependent haemodynamic behaviour (including flow deceleration and oxygen exchange) similar to rodents and can be analysed in the same spectrum as rodents. Finally, we show strong structure-function relationships when interspecies datasets are combined; notably, we demonstrate that surrogate measures such as vascularity, can be used to estimate placental oxygen exchange function. DISCUSSION Pre-clinical placental research utilising rat and mouse placentae to understand the impact of feto-placental arborization on placental function and fetal development can inform the human context.
Collapse
Affiliation(s)
- Nikhilesh Bappoo
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia; School of Human Sciences, The University of Western Australia, Perth, Australia
| | - Yutthapong Tongpob
- School of Human Sciences, The University of Western Australia, Perth, Australia; Centre of Excellence in Medical Biotechnology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Matina Hakim
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, UK
| | - Jenny Myers
- Maternal and Fetal Health Research Centre, Division of Developmental Biology and Medicine, University of Manchester, Manchester, UK
| | - Emma Panting
- School of Human Sciences, The University of Western Australia, Perth, Australia
| | - Karen E Chapman
- School of Human Sciences, The University of Western Australia, Perth, Australia; Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Adrian J W Thomson
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Carmel M Moran
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, EH16 4TJ, UK
| | - Lachlan J Kelsey
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia
| | | | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Alys R Clark
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Barry J Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia, Perth, Australia; School of Engineering, The University of Western Australia, Perth, Australia
| | - Caitlin S Wyrwoll
- School of Human Sciences, The University of Western Australia, Perth, Australia; Telethon Kids Institute, Perth, Australia.
| |
Collapse
|
3
|
Saghian R, Cahill LS, Debebe SK, Rahman A, Serghides L, McDonald CR, Weckman AM, Kain KC, Sled JG. Allometric scaling relationships in mouse placenta. J R Soc Interface 2022; 19:20220579. [PMID: 36349448 PMCID: PMC9653247 DOI: 10.1098/rsif.2022.0579] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/19/2022] [Indexed: 08/29/2023] Open
Abstract
Fetal growth and maturation are highly intertwined with placental development during pregnancy. Here we used placental vascular morphology measurements (depth and span) as well as the umbilical artery (UA) diameter of previously published studies on three different mouse strains (C57BL6/J, CD-1 and BALB/c), which were exposed to different conditions (combination antiretroviral therapy, chronic maternal hypoxia and malaria infection) at different embryonic days, to test the hypothesis that placental vascularization and specifically the UA size affect conceptus weight. Interaction of each study parameter with embryonic day, strain and exposure to treatments are studied to investigate the stability of the scaling relationships across and/or within strains and conditions. In addition, the effect of UA diameter on the placental growth measurements (depth and span) is studied. These results show that the power-law scaling relationship of conceptus weight and placental depth with the UA diameter is conserved across strains and conditions with the scaling exponent of approximately 3/8 and 5/8, respectively. By contrast, the relationship between conceptus weight and either the placental span or depth is different between strains and conditions, suggesting multiple mechanisms of vascular adaptation.
Collapse
Affiliation(s)
- Rojan Saghian
- Mouse Imaging Centre, 25 Orde Street, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lindsay S. Cahill
- Mouse Imaging Centre, 25 Orde Street, Toronto, Ontario, Canada
- Department of Chemistry, Memorial University of Newfoundland, Newfoundland and Labrador, St John’s, Canada
| | - Sarah K. Debebe
- Mouse Imaging Centre, 25 Orde Street, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
| | - Anum Rahman
- Mouse Imaging Centre, 25 Orde Street, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
| | - Lena Serghides
- Department of Immunology and Institute of Medical Sciences, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
- Women’s College Research Institute, Women’s College Hospital, Toronto, Ontario, Canada
| | - Chloe R. McDonald
- Institute of Medical Science, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
| | - Andrea M. Weckman
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Kevin C. Kain
- Institute of Medical Science, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - John G. Sled
- Mouse Imaging Centre, 25 Orde Street, Toronto, Ontario, Canada
- Translational Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Biophysics, University Health Network-Toronto General Hospital, Toronto, Ontario, Canada
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
4
|
Liu XC, Strodl E, Wu CA, Huang LH, Yin XN, Wen GM, Sun DL, Xian DX, Chen WQ. Critical window for the association between prenatal environmental tobacco smoke exposure and preterm birth. ENVIRONMENTAL RESEARCH 2022; 212:113427. [PMID: 35561826 DOI: 10.1016/j.envres.2022.113427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 06/15/2023]
Abstract
Although environmental tobacco smoke (ETS) exposure is considered to be a severe public health problem and a modifiable risk factor for preterm birth (PTB), we still lack a comprehensive understanding of the PTB risk associated with trimester-specific prenatal ETS exposure. This study aimed to examine the accumulation of risk across trimester ETS exposure and the critical window of the association between maternal ETS exposure during pregnancy and PTB. A total of 63,038 mother-child pairs were involved in the analysis of the 2017 survey of Longhua Child Cohort Study. Information about socio-demographic characteristics, prenatal ETS exposure, and birth outcomes were collected using a self-report questionnaire. A series of logistic regression models were employed to assess the associations between prenatal ETS exposure and PTB. We found that maternal ETS exposure during pregnancy significantly increased the risk of PTB and this association increased with both the average level of daily ETS exposure and the number of trimesters of ETS exposure. Moreover, mothers who were initially exposed to ETS in the 1st trimester of pregnancy had significant higher risk of PTB (OR = 1.34, 95% CI: 1.25-1.44). Furthermore, mothers exposed to ETS in the 1st trimester only (OR = 1.26, 95%CI: 1.04-1.50), in both 1st and 2nd trimester (OR = 1.35, 95%CI: 1.08-1.67) and throughout pregnancy (OR = 1.35, 95%CI: 1.24-1.46) experienced a significantly high risk of PTB. Prenatal maternal ETS exposure during only the 2nd trimester also resulted in a high risk of PTB with marginal significance (OR = 1.33, 95% CI:0.78-2.13). To conclude, the 1st and early 2nd trimester might be the critical window for prenatal ETS exposure causing PTB.
Collapse
Affiliation(s)
- Xin-Chen Liu
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Esben Strodl
- School of Psychology and Counselling, Queensland University of Technology, Brisbane, QLD, Australia
| | - Chuan-An Wu
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, China
| | - Li-Hua Huang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Xiao-Na Yin
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, China
| | - Guo-Min Wen
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, China
| | - Deng-Li Sun
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, China
| | - Dan-Xia Xian
- Women's and Children's Hospital of Longhua District of Shenzhen, Shenzhen, China
| | - Wei-Qing Chen
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China; School of Health, Xinhua College of Guangzhou, China.
| |
Collapse
|
5
|
Aghaei Z, Steeves KL, Jobst KJ, Cahill LS. The impact of perfluoroalkyl substances on pregnancy, birth outcomes and offspring development: A review of data from mouse models1. Biol Reprod 2021; 106:397-407. [PMID: 34875017 DOI: 10.1093/biolre/ioab223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 11/12/2022] Open
Abstract
Per- and polyfluoroalkyl substances (PFASs) such as perfluorooctanoic acid (PFOA) and perfluorooctanesulfonic acid (PFOS) are persistent in the environment and bioaccumulate in wildlife and humans, potentially causing adverse health effects at all stages of life. Studies from human pregnancy have shown that exposure to these contaminants are associated with placental dysfunction and fetal growth restriction; however, studies in humans are confounded by genetic and environmental factors. Here, we synthesize the available results from mouse models of pregnancy to show the causal effects of prenatal exposure to PFOA and PFOS on placental and fetal development and on neurocognitive function and metabolic disorders in offspring. We also propose gaps in the present knowledge and provide suggestions for future research studies.
Collapse
Affiliation(s)
- Zahra Aghaei
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Katherine L Steeves
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Karl J Jobst
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Lindsay S Cahill
- Department of Chemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
6
|
Tran V, Weckman AM, Crowley VM, Cahill LS, Zhong K, Cabrera A, Elphinstone RE, Pearce V, Madanitsa M, Kalilani-Phiri L, Mwapasa V, Khairallah C, Conroy AL, Ter Kuile FO, Sled JG, Kain KC. The Angiopoietin-Tie2 axis contributes to placental vascular disruption and adverse birth outcomes in malaria in pregnancy. EBioMedicine 2021; 73:103683. [PMID: 34758414 PMCID: PMC8590041 DOI: 10.1016/j.ebiom.2021.103683] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/28/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022] Open
Abstract
Background Malaria during pregnancy is a major contributor to the global burden of adverse birth outcomes including fetal growth restriction, preterm birth, and fetal loss. Recent evidence supports a role for angiogenic dysregulation and perturbations to placental vascular development in the pathobiology of malaria in pregnancy. The Angiopoietin-Tie2 axis is critical for placental vascularization and remodeling. We hypothesized that disruption of this pathway would contribute to malaria-induced adverse birth outcomes. Methods Using samples from a previously conducted prospective cohort study of pregnant women in Malawi, we measured circulating levels of angiopoietin-1 (Angpt-1) and Angpt-2 by Luminex (n=1392). We used a preclinical model of malaria in pregnancy (Plasmodium berghei ANKA [PbA] in pregnant BALB/c mice), genetic disruption of Angpt-1 (Angpt1+/− mice), and micro-CT analysis of placental vasculature to test the hypothesis that disruptions to the Angpt-Tie2 axis by malaria during pregnancy would result in aberrant placental vasculature and adverse birth outcomes. Findings Decreased circulating levels of Angpt-1 and an increased ratio of Angpt-2/Angpt-1 across pregnancy were associated with malaria in pregnancy. In the preclinical model, PbA infection recapitulated disruptions to the Angiopoietin-Tie2 axis resulting in reduced fetal growth and viability. Malaria decreased placental Angpt-1 and Tie2 expression and acted synergistically with reduced Angpt-1 in heterozygous dams (Angpt1+/−), to worsen birth outcomes by impeding vascular remodeling required for placental function. Interpretation Collectively, these data support a mechanistic role for the Angpt-Tie2 axis in malaria in pregnancy, including a potential protective role for Angpt-1 in mitigating infection-associated adverse birth outcomes. Funding This work was supported by the Canadian Institutes of Health Research (CIHR), Canada Research Chair, and Toronto General Research Institute Postdoctoral Fellowship Award. The parent trial was supported by the European & Developing Countries Clinical Trials Partnership and the Malaria in Pregnancy Consortium, which was funded by the Bill & Melinda Gates Foundation. The funders had no role in design, analysis, or reporting of these studies.
Collapse
Affiliation(s)
- Vanessa Tran
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada
| | - Andrea M Weckman
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada
| | - Valerie M Crowley
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Canada
| | - Kathleen Zhong
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada
| | - Ana Cabrera
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, Canada
| | - Robyn E Elphinstone
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada
| | - Victoria Pearce
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada
| | - Mwayiwawo Madanitsa
- Department of Clinical Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Thyolo, Malawi
| | | | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Carole Khairallah
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Andrea L Conroy
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, United States
| | - Feiko O Ter Kuile
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Canada
| | - Kevin C Kain
- SAR Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital Research Institute, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Canada; Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
7
|
Bappoo N, Kelsey LJ, Tongpob Y, Wyrwoll C, Doyle BJ. Investigating the Upstream and Downstream Hemodynamic Boundary Conditions of Healthy and Growth-Restricted Rat Feto-Placental Arterial Networks. Ann Biomed Eng 2021; 49:2183-2195. [PMID: 33646497 DOI: 10.1007/s10439-021-02749-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 02/05/2021] [Indexed: 11/30/2022]
Abstract
The placenta uniquely develops to orchestrate maternal adaptations and support fetal growth and development. The expansion of the feto-placental vascular network, in part, underpins function. However it is unclear how vascular development is synergistically influenced by hemodynamics and how impairment may lead to fetal growth restriction (FGR). Here, we present a robust framework consisting of ex vivo placental casting, imaging and computational fluid dynamics of rat feto-placental networks where we investigate inlet (steady and transient) and outlet (zero-pressure, Murray's Law, asymmetric fractal trees and porous blocks) boundary conditions in a model of growth-restriction. We show that the Murray's Law flow-split boundary condition is not always appropriate and that mean steady-state inlet conditions produce comparable results to transient flow. However, we conclude that transient simulations should be adopted as they provide a larger amount of valuable data, a necessity to bridge the current knowledge gap in placental biomechanics. We also show preliminary data on changes in flow, shear stress, and flow deceleration between control and growth-restricted feto-placental networks. Our proposed framework provides a standardized approach for structural and hemodynamic analysis of feto-placental vasculature and has the potential to enhance our understanding of placental function.
Collapse
Affiliation(s)
- Nikhilesh Bappoo
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, WA, 6009, Australia.
- School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia.
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia.
| | - Lachlan J Kelsey
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, WA, 6009, Australia
- School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
| | - Yutthapong Tongpob
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
- Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Caitlin Wyrwoll
- School of Human Sciences, The University of Western Australia, Perth, WA, 6009, Australia
| | - Barry J Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, Nedlands, WA, 6009, Australia
- School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, Australia
- BHF Centre of Cardiovascular Science, The University of Edinburgh, Edinburgh, EH9 3FD, UK
| |
Collapse
|
8
|
James JL, Tongpob Y, Srinivasan V, Crew RC, Bappoo N, Doyle B, Gerneke D, Clark AR, Wyrwoll CS. Three-dimensional visualisation of the feto-placental vasculature in humans and rodents. Placenta 2021; 114:8-13. [PMID: 34418753 DOI: 10.1016/j.placenta.2021.08.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/09/2021] [Accepted: 08/12/2021] [Indexed: 11/19/2022]
Abstract
Adequate development of the feto-placental circulation is critical for placental exchange function and healthy fetal growth. Understanding the structure of this circulation and how it informs fetal outcomes is important both in the human placenta, and the rodent, a purported comparative experimental model. Vascular casting and micro-CT imaging approaches enable detailed quantification of the complex vascular relationships in the feto-circulation, and provide detailed data to parameterise in silico models. Here, to assist researchers to apply these technically challenging methods we provide detailed approaches to cast and image; 1) human placentas at the cotyledon-level, and 2) whole rodent placentas.
Collapse
Affiliation(s)
- J L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Y Tongpob
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - V Srinivasan
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - R C Crew
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - N Bappoo
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia; Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, WA, Australia; School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia
| | - B Doyle
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and the UWA Centre for Medical Research, The University of Western Australia, WA, Australia; School of Engineering, The University of Western Australia, Perth, WA, 6009, Australia; Australian Research Council Centre for Personalised Therapeutics Technologies, Australia; Centre for Cardiovascular Science, The University of Edinburgh, UK
| | - D Gerneke
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - A R Clark
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - C S Wyrwoll
- School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
9
|
Saghian R, Cahill L, Rahman A, Steinman J, Stortz G, Kingdom J, Macgowan C, Sled J. Interpretation of wave reflections in the umbilical arterial segment of the feto-placental circulation: computational modeling of the feto-placental arterial tree. IEEE Trans Biomed Eng 2021; 68:3647-3658. [PMID: 34010124 DOI: 10.1109/tbme.2021.3082064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Placental vascular abnormalities are associated with a host of pregnancy complications including placenta mediated fetal growth restriction (FGR). Umbilical arterial (UA) Doppler ultrasound velocity waveforms are widely used in the diagnosis of underlying placental vascular abnormalities in pregnancies with suspected FGR, which greatly help prevent stillbirth via ongoing fetal monitoring and timely delivery. However, the sensitivity of UA Doppler diagnosis diminishes late in gestation. Our goal was to present a generalized wave decomposition method to compute forward and reflected components from UA waveforms. A detailed anatomical based model was also developed to explain observed UA flow waveform and to explore how vascular properties affect the shape of flow wave components. Using pregnant mice and high frequency ultrasound microscopy, we obtained in utero Doppler and M- mode ultrasound measurements in 15 fetuses UA. Following ultrasound, the placentas were collected and perfused with contrast agent to obtain high-resolution 3D images of the feto-placental arteries. Model results indicate the significant role of terminal load impedance (capillary and/or veins) in creating positive or negative reflected waveforms. A negative reflected waveform is obtained when terminal impedance increases. This is consistent with the elongated and non-branching terminal villi that are proposed cause the highly abnormal UA waveforms found in early-onset FGR. The significance of these findings for the diagnostic utility of UA Doppler in human pregnancy is that the identification and measurement of wave reflections may aid in discriminating between healthy and abnormal placental vasculature in pregnancies with suspected late-onset FGR.
Collapse
|
10
|
Byrne M, Aughwane R, James JL, Hutchinson JC, Arthurs OJ, Sebire NJ, Ourselin S, David AL, Melbourne A, Clark AR. Structure-function relationships in the feto-placental circulation from in silico interpretation of micro-CT vascular structures. J Theor Biol 2021; 517:110630. [PMID: 33607145 DOI: 10.1016/j.jtbi.2021.110630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/28/2021] [Accepted: 02/08/2021] [Indexed: 10/22/2022]
Abstract
A well-functioning placenta is critical for healthy fetal development, as the placenta brings fetal blood in close contact with nutrient rich maternal blood, enabling exchange of nutrients and waste between mother and fetus. The feto-placental circulation forms a complex branching structure, providing blood to fetal capillaries, which must receive sufficient blood flow to ensure effective exchange, but at a low enough pressure to prevent damage to placental circulatory structures. The branching structure of the feto-placental circulation is known to be altered in complications such as fetal growth restriction, and the presence of regions of vascular dysfunction (such as hypovascularity or thrombosis) are proposed to elevate risk of placental pathology. Here we present a methodology to combine micro-computed tomography and computational model-based analysis of the branching structure of the feto-placental circulation in ex vivo placentae from normal term pregnancies. We analyse how vascular structure relates to function in this key organ of pregnancy; demonstrating that there is a 'resilience' to placental vascular structure-function relationships. We find that placentae with variable chorionic vascular structures, both with and without a Hyrtl's anastomosis between the umbilical arteries, and those with multiple regions of poorly vascularised tissue are able to function with a normal vascular resistance. Our models also predict that by progressively introducing local heterogeneity in placental vascular structure, large increases in feto-placental vascular resistances are induced. This suggests that localised heterogeneities in placental structure could potentially provide an indicator of increased risk of placental dysfunction.
Collapse
Affiliation(s)
- Monika Byrne
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Rosalind Aughwane
- Department of Maternal Fetal Medicine, Prenatal Cell and Gene Therapy Group, Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, WC1E 6HX, United Kingdom
| | - Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - J Ciaran Hutchinson
- NIHR GOS Institute of Child Health Biomedical Research Centre, University College, London, United Kingdom; Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Owen J Arthurs
- NIHR GOS Institute of Child Health Biomedical Research Centre, University College, London, United Kingdom; Paediatric Radiology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Neil J Sebire
- NIHR GOS Institute of Child Health Biomedical Research Centre, University College, London, United Kingdom; Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Sebastien Ourselin
- School of Biomedical Engineering and Imaging Sciences, Kings College London, United Kingdom
| | - Anna L David
- Department of Maternal Fetal Medicine, Prenatal Cell and Gene Therapy Group, Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, WC1E 6HX, United Kingdom; NIHR University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road, London, W1T 7DN, United Kingdom
| | - Andrew Melbourne
- School of Biomedical Engineering and Imaging Sciences, Kings College London, United Kingdom
| | - Alys R Clark
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
11
|
Flouri D, Darby JRT, Holman SL, Perumal SR, David AL, Morrison JL, Melbourne A. Magnetic resonance imaging of placentome development in the pregnant Ewe. Placenta 2021; 105:61-69. [PMID: 33549925 PMCID: PMC7611430 DOI: 10.1016/j.placenta.2021.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Novel imaging measurements of placental development are difficult to validate due to the invasive nature of gold-standard procedures. Animal studies have been important in validation of magnetic resonance imaging (MRI) measurements in invasive preclinical studies, as they allow for controlled experiments and analysis of multiple time-points during pregnancy. This study characterises the longitudinal diffusion and perfusion properties of sheep placentomes using MRI, measurements that are required for future validation studies. METHODS Pregnant ewes were anaesthetised for a MRI session on a 3T scanner. Placental MRI was used to classify placentomes morphologically into three types based on their shape and size at two gestational ages. To validate classification accuracy, placentome type derived from MRI data were compared with placentome categorisation results after delivery. Diffusion-Weighted MRI and T2-relaxometry were used to measure a broad range of biophysical properties of the placentomes. RESULTS MRI morphological classification results showed consistent gestational age changes in placentome shape, as supported by post-delivery gold standard data. The mean apparent diffusion coefficient was significantly higher at 110 days gestation than at late gestation (~140 days; term, 150 days). Mean T2 was higher at mid gestation (152.2 ± 58.1 ms) compared to late gestation (127.8 ms ± 52.0). Significantly higher perfusion fraction was measured in late gestation placentomes that also had a significantly higher fractional anisotropy when compared to the earlier gestational age. DISCUSSION We report baseline measurements of techniques common in placental MRI for the sheep placenta. These measurements are essential to support future validation measurements of placental MRI techniques.
Collapse
Affiliation(s)
- Dimitra Flouri
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, United Kingdom; Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom.
| | - Jack R T Darby
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Stacey L Holman
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Sunthara R Perumal
- South Australian Health & Medical Research Institute, Preclinical, Imaging & Research Laboratories, Adelaide, Australia
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, London, London, United Kingdom; NIHR Biomedical Research Centre, University College London Hospitals, London, United Kingdom
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Andrew Melbourne
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, United Kingdom; Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom; Early Origins of Adult Health Research Group, Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| |
Collapse
|
12
|
Perera-Bel E, Ceresa M, Torrents-Barrena J, Masoller N, Valenzuela-Alcaraz B, Gratacós E, Eixarch E, González Ballester MA. Segmentation of the placenta and its vascular tree in Doppler ultrasound for fetal surgery planning. Int J Comput Assist Radiol Surg 2020; 15:1869-1879. [PMID: 32951100 DOI: 10.1007/s11548-020-02256-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 09/02/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE Twin-to-twin transfusion syndrome (TTTS) is a serious condition that occurs in about 10-15% of monochorionic twin pregnancies. In most instances, the blood flow is unevenly distributed throughout the placenta anastomoses leading to the death of both fetuses if no surgical procedure is performed. Fetoscopic laser coagulation is the optimal therapy to considerably improve co-twin prognosis by clogging the abnormal anastomoses. Notwithstanding progress in recent years, TTTS surgery is highly risky. Computer-assisted planning of the intervention can thus improve the outcome. METHODS In this work, we implement a GPU-accelerated random walker (RW) algorithm to detect the placenta, both umbilical cord insertions and the placental vasculature from Doppler ultrasound (US). Placenta and background seeds are manually initialized in 10-20 slices (out of 245). Vessels are automatically initialized in the same slices by means of Otsu thresholding. The RW finds the boundaries of the placenta and reconstructs the vasculature. RESULTS We evaluate our semiautomatic method in 5 monochorionic and 24 singleton pregnancies. Although satisfactory performance is achieved on placenta segmentation (Dice ≥ 84.0%), some vascular connections are still neglected due to the presence of US reverberation artifacts (Dice ≥ 56.9%). We also compared inter-user variability and obtained Dice coefficients of ≥ 76.8% and ≥ 97.42% for placenta and vasculature, respectively. After a 3-min manual initialization, our GPU approach speeds the computation 10.6 times compared to the CPU. CONCLUSIONS Our semiautomatic method provides a near real-time user experience and requires short training without compromising the segmentation accuracy. A powerful approach is thus presented to rapidly plan the fetoscope insertion point ahead of TTTS surgery.
Collapse
Affiliation(s)
- Enric Perera-Bel
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain.
| | - Mario Ceresa
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Jordina Torrents-Barrena
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Narcís Masoller
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Brenda Valenzuela-Alcaraz
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Eduard Gratacós
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Elisenda Eixarch
- BCNatal, Fetal Medicine Research Center (Hospital Clínic and Hospital Sant Joan de Déu), University of Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centre for Biomedical Research on Rare Diseases (CIBER-ER), Madrid, Spain
| | - Miguel A González Ballester
- Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain.,ICREA, Barcelona, Spain
| |
Collapse
|
13
|
Maliszewska-Cyna E, Vecchio LM, Thomason LAM, Oore JJ, Steinman J, Joo IL, Dorr A, McLaurin J, Sled JG, Stefanovic B, Aubert I. The effects of voluntary running on cerebrovascular morphology and spatial short-term memory in a mouse model of amyloidosis. Neuroimage 2020; 222:117269. [PMID: 32818618 DOI: 10.1016/j.neuroimage.2020.117269] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/06/2020] [Accepted: 08/10/2020] [Indexed: 12/27/2022] Open
Abstract
Physical activity has been correlated with a reduced risk of cognitive decline, including that associated with vascular dementia, mild cognitive impairment (MCI) and Alzheimer's disease (AD); recent literature suggests this may in part result from benefits to the cerebrovascular network. Using a transgenic (Tg) mouse model of AD, we evaluated the effect of running on cortical and hippocampal vascular morphology, cerebral amyloid angiopathy, amyloid plaque load, and spatial memory. TgCRND8 mice present with progressive amyloid pathology, advancing from the cortex to the hippocampus in a time-dependent manner. We postulated that the characteristic progression of pathology could lead to differential, time-dependent effects of physical activity on vascular morphology in these brain regions at 6 months of age. We used two-photon fluorescent microscopy and 3D vessel tracking to characterize vascular and amyloid pathology in sedentary TgCRND8 mice compared those who have a history of physical activity (unlimited access to a running wheel, from 3 to 6 months of age). In sedentary TgCRND8 mice, capillary density was found to be lower in the cortex and higher in the hippocampus compared to non-transgenic (nonTg) littermates. Capillary length, vessel branching, and non-capillary vessel tortuosity were also higher in the hippocampus of sedentary TgCRND8 compared to nonTg mice. Three months of voluntary running resulted in normalizing cortical and hippocampal microvascular morphology, with no significant difference between TgCRND8 and nonTg mice. The benefits of physical activity on cortical and hippocampal vasculature in 6-month old TgCRND8 mice were not paralleled by significant changes on parenchymal and cerebral amyloid pathology. Short-term spatial memory- as evaluated by performance in the Y-maze- was significantly improved in running compared to sedentary TgCRND8 mice. These results suggest that long-term voluntary running contributes to the maintenance of vascular morphology and spatial memory in TgCRND8 mice, even in the absence of an effect on amyloid pathology.
Collapse
Affiliation(s)
- Ewelina Maliszewska-Cyna
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Laura M Vecchio
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.
| | - Lynsie A M Thomason
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - Jonathan J Oore
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada
| | - Joe Steinman
- Mouse Imaging Centre, Hospital for Sick Children, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Illsung Lewis Joo
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - Adrienne Dorr
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada
| | - JoAnne McLaurin
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Bojana Stefanovic
- Hurvitz Brain Sciences, Physical Sciences, Sunnybrook Research Institute, Canada; Department of Medical Biophysics, University of Toronto, Canada
| | - Isabelle Aubert
- Hurvitz Brain Sciences, Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S112, Toronto, Ontario M4N 3M5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
14
|
Advances in imaging feto-placental vasculature: new tools to elucidate the early life origins of health and disease. J Dev Orig Health Dis 2020; 12:168-178. [PMID: 32746961 DOI: 10.1017/s2040174420000720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Optimal placental function is critical for fetal development, and therefore a crucial consideration for understanding the developmental origins of health and disease (DOHaD). The structure of the fetal side of the placental vasculature is an important determinant of fetal growth and cardiovascular development. There are several imaging modalities for assessing feto-placental structure including stereology, electron microscopy, confocal microscopy, micro-computed tomography, light-sheet microscopy, ultrasonography and magnetic resonance imaging. In this review, we present current methodologies for imaging feto-placental vasculature morphology ex vivo and in vivo in human and experimental models, their advantages and limitations and how these provide insight into placental function and fetal outcomes. These imaging approaches add important perspective to our understanding of placental biology and have potential to be new tools to elucidate a deeper understanding of DOHaD.
Collapse
|
15
|
Reijnders IF, Mulders AGMGJ, van der Windt M, Steegers EAP, Steegers-Theunissen RPM. The impact of periconceptional maternal lifestyle on clinical features and biomarkers of placental development and function: a systematic review. Hum Reprod Update 2020; 25:72-94. [PMID: 30407510 DOI: 10.1093/humupd/dmy037] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/17/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Worldwide, placenta-related complications contribute to adverse pregnancy outcomes, such as pre-eclampsia, fetal growth restriction and preterm birth, with implications for the future health of mothers and offspring. The placenta develops in the periconception period and forms the interface between mother and embryo/fetus. An unhealthy periconceptional maternal lifestyle, such as smoking, alcohol and under- and over-nutrition, can detrimentally influence placental development and function. OBJECTIVE AND RATIONALE The impact of maternal lifestyle on placental health is largely unknown. Therefore, we aim to summarize the evidence of the impact of periconceptional maternal lifestyle on clinical features and biomarkers of placental development and function throughout pregnancy. SEARCH METHODS A comprehensive search in Medline, Embase, Pubmed, The Cochrane Library Web of Science and Google Scholar was conducted. The search strategy included keywords related to the maternal lifestyle, smoking, alcohol, caffeine, nutrition (including folic acid supplement intake) and body weight. For placental markers throughout pregnancy, keywords related to ultrasound imaging, serum biomarkers and histological characteristics were used. We included randomized controlled trials and observational studies published between January 2000 and March 2017 and restricted the analysis to singleton pregnancies and maternal periconceptional lifestyle. Methodological quality was scored using the ErasmusAGE tool. A protocol of this systematic review has been registered in PROSPERO International prospective register of systematic reviews (PROSPERO 2016:CRD42016045596). OUTCOMES Of 2593 unique citations found, 82 studies were included. The median quality score was 5 (range: 0-10). The findings revealed that maternal smoking was associated with lower first-trimester placental vascularization flow indices, higher second- and third-trimester resistance of the uterine and umbilical arteries and lower resistance of the middle cerebral artery. Although a negative impact of smoking on placental weight was expected, this was less clear. Alcohol use was associated with a lower placental weight. One study described higher second- and third-trimester placental growth factor (PlGF) levels after periconceptional alcohol use. None of the studies looked at caffeine intake. Adequate nutrition in the first trimester, periconceptional folic acid supplement intake and strong adherence to a Mediterranean diet, were all associated with a lower resistance of the uterine and umbilical arteries in the second and third trimester. A low caloric intake resulted in a lower placental weight, length, breadth, thickness, area and volume. Higher maternal body weight was associated with a larger placenta measured by ultrasound in the second and third trimester of pregnancy or weighed at birth. In addition, higher maternal body weight was associated with decreased PlGF-levels. WIDER IMPLICATIONS Evidence of the impact of periconceptional maternal lifestyle on placental health was demonstrated. However, due to poorly defined lifestyle exposures and time windows of investigation, unstandardized measurements of placenta-related outcomes and small sample sizes of the included studies, a cautious interpretation of the effect estimates is indicated. We suggest that future research should focus more on physiological consequences of unhealthy lifestyle during the critical periconception window. Moreover, we foresee that new evidence will support the development of lifestyle interventions to improve the health of mothers and their offspring from the earliest moment in life.
Collapse
Affiliation(s)
- Ignatia F Reijnders
- Department of Obstetrics and Gynaecology, Erasmus Medical Centre, University Medical Centre, Rotterdam, The Netherlands
| | - Annemarie G M G J Mulders
- Department of Obstetrics and Gynaecology, Erasmus Medical Centre, University Medical Centre, Rotterdam, The Netherlands
| | - Melissa van der Windt
- Department of Obstetrics and Gynaecology, Erasmus Medical Centre, University Medical Centre, Rotterdam, The Netherlands
| | - Eric A P Steegers
- Department of Obstetrics and Gynaecology, Erasmus Medical Centre, University Medical Centre, Rotterdam, The Netherlands
| | | |
Collapse
|
16
|
Effects of maternal smoking on human placental vascularization: A systematic review. Taiwan J Obstet Gynecol 2020; 58:454-459. [PMID: 31307732 DOI: 10.1016/j.tjog.2019.05.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2019] [Indexed: 12/11/2022] Open
Abstract
The abnormal development of placental vascularization leads to placental insufficiency, which further reduces the nutrient and trace exchange between maternal circulation and fetal circulation. These changes cause maternal and fetal complications. The objective of our systematic review was to explore the effects of maternal smoking on placental vascularization. The eligibility criteria were: articles with experimental, quasi-experimental or observational design, performed on human subjects, that study the association, correlation or causation between maternal smoking and changes in placental vascular network. A total of 33 full-text papers were assessed for eligibility, resulting in 12 original articles that were included in the systematic review. Doppler studies confirm reductions in blood flow velocity waveforms and increase in RI in the uterine, umbilical and fetal middle cerebral arteries. These findings are confirmed by morphometric measurements of fetal capillaries in villi that were shown to be smaller in smoke exposure groups.
Collapse
|
17
|
Acute and chronic stage adaptations of vascular architecture and cerebral blood flow in a mouse model of TBI. Neuroimage 2019; 202:116101. [DOI: 10.1016/j.neuroimage.2019.116101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 11/18/2022] Open
|
18
|
Tongpob Y, Xia S, Wyrwoll C, Mehnert A. Quantitative characterization of rodent feto-placental vasculature morphology in micro-computed tomography images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 179:104984. [PMID: 31443859 DOI: 10.1016/j.cmpb.2019.104984] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 06/18/2019] [Accepted: 07/13/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVE Optimal development of placental vasculature is critical for fetal growth and health outcomes. Many studies characterizing the vascular structure of the fetal side of the placenta have utilized a range of two-dimensional and three-dimensional (3D) imaging techniques including X-ray micro-computed tomography (micro-CT) following perfusion of the vasculature with a radio-opaque compound. The CT approach has been used to study feto-placental vasculature in rodents and humans. Its inherent advantage is that it reveals the 3D structure in high resolution without destroying the sample. This permits both multiple scanning of the sample and follow-up histological investigations in the same sample. Nevertheless, the applicability of the approach is hampered both by the challenging segmentation of the vasculature and a lack of straightforward methodology to quantitate the feto-placental vascular network. This paper addresses these challenges. METHODS An end-to-end methodology is presented for automatically segmenting the vasculature; obtaining a Strahler-ordered rooted-tree representation and extracting quantitative features from its nodes, segments and branches (including volume, length, tortuosity and branching angles). The methodology is demonstrated for rat and mouse placentas at the end of gestation (day 22 and day 18, respectively), perfused with Microfil® and imaged using two different micro-CT scanners. RESULTS The 3D visualizations of the resulting vascular trees clearly demonstrate differences between the branching complexity, tree span and tree depth of the mouse and rat placentas. The quantitative characterizations of these trees include not only the fundamental features that have been utilized in other studies of feto-placental vasculature but also several additional features. Boxplots of several of these-tortuosity, number of side branches, number of offspring per branch and branch volume-computed at each Strahler order are presented and interpreted. Differences and similarities between the mouse and rat casts are readily detected. CONCLUSION The proposed end-to-end methodology, and the implementation presented using a combination of Amira and Matlab, offers researchers in the field of placental vasculature characterization a straightforward and objective approach for quantifying micro-CT vascular datasets.
Collapse
Affiliation(s)
- Yutthapong Tongpob
- School of Human Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia; Faculty of Medical Science, Naresuan University, 99 Moo 9, Tapo, Muang, Phitsanulok 65000, Thailand.
| | - Shushan Xia
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, 306, N Wright St, Urbana, IL 61801, USA; Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia
| | - Caitlin Wyrwoll
- School of Human Sciences, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia.
| | - Andrew Mehnert
- Centre for Microscopy, Characterisation and Analysis, The University of Western Australia, 35 Stirling Hwy, Crawley, Perth, WA 6009, Australia; National Imaging Facility, Western Australian Node, The University of Western Australia, Perth, WA 6009, Australia.
| |
Collapse
|
19
|
Micro-CT and histological investigation of the spatial pattern of feto-placental vascular density. Placenta 2019; 88:36-43. [PMID: 31670095 PMCID: PMC6892277 DOI: 10.1016/j.placenta.2019.09.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 09/18/2019] [Accepted: 09/27/2019] [Indexed: 12/03/2022]
Abstract
Introduction There are considerable variations in villous morphology within a normal placenta. However, whether there is a reproducible spatial pattern of variation in villous vascular density is not known. Micro-CT provides three-dimensional volume imaging with spatial resolution down to the micrometre scale. In this study, we applied Micro-CT and histological analysis to investigate the degree of heterogeneity of vascularisation within the placenta. Method Ten term placentas were collected at elective caesarean section, perfused with contrast agent and imaged whole with Micro-CT. Eight full depth tissue blocks were then taken from each placenta and imaged. Sections were taken for histological analysis. Data was analysed to investigate vascular fill, and vascular density in relation to location from cord insertion to placental edge at each scale. Results Whole placental imaging revealed no spatially consistent difference in villous vessel density within the main placental tissue, although there was a great degree of heterogeneity. Both block imaging and histological analysis found a large degree of heterogeneity of vascular density within placentas, but no strong correlation between villous vascular density and block location (rs = 0.066, p = 0.7 block imaging, rs = 0.06, p = 0.6 histological analysis). Discussion This work presents a novel method for imaging the human placenta vascular tree using multiscale Micro-CT imaging. It demonstrates that there is a large degree of variation in vascular density throughout normal term human placentas. The three-dimensional data created by this technique could be used, with more advanced computer analysis, to further investigate the structure of the vascular tree. Micro-CT and histological investigation of vascular density in the placenta. There is a large degree of variation in vascular density throughout placentas. This imaging has potential for future spatial investigation of the 3D vascular tree.
Collapse
|
20
|
McDonald CR, Cahill LS, Gamble JL, Elphinstone R, Gazdzinski LM, Zhong KJY, Philson AC, Madanitsa M, Kalilani-Phiri L, Mwapasa V, Ter Kuile FO, Sled JG, Conroy AL, Kain KC. Malaria in pregnancy alters l-arginine bioavailability and placental vascular development. Sci Transl Med 2019. [PMID: 29514999 PMCID: PMC6510298 DOI: 10.1126/scitranslmed.aan6007] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Reducing adverse birth outcomes due to malaria in pregnancy (MIP) is a global health priority. However, there are few safe and effective interventions. L-arginine is an essential amino acid in pregnancy and an immediate precursor in the biosynthesis of nitric oxide (NO), but there are limited data on the impact of MIP on NO biogenesis. We hypothesized that hypoarginemia contributes to the pathophysiology of MIP and that L-arginine supplementation would improve birth outcomes. In a prospective study of pregnant Malawian women, we show that MIP was associated with lower concentrations of L- arginine and higher concentrations of endogenous inhibitors of NO biosynthesis, asymmetric and symmetric dimethylarginine, which were associated with adverse birth outcomes. In a model of experimental MIP, L-arginine supplementation in dams improved birth outcomes (decreased stillbirth and increased birth weight) compared with controls. The mechanism of action was via normalized angiogenic pathways and enhanced placental vascular development, as visualized by placental microcomputerized tomography imaging. These data define a role for dysregulation of NO biosynthetic pathways in the pathogenesis of MIP and support the evaluation of interventions to enhance L-arginine bioavailability as strategies to improve birth outcomes.
Collapse
Affiliation(s)
- Chloe R McDonald
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Joel L Gamble
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Robyn Elphinstone
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Lisa M Gazdzinski
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Kathleen J Y Zhong
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Adrienne C Philson
- Howard Hughes Medical Institute, Department of Neurobiology, Harvard Medical School, Boston, MA 02138, USA
| | | | | | - Victor Mwapasa
- College of Medicine, University of Malawi, P.O. Box 280, Blantyre, Malawi
| | | | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario M5T 3HT, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 1L7, Canada
| | - Andrea L Conroy
- Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| | - Kevin C Kain
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada. .,Sandra A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, University of Toronto, Toronto, Ontario M5G 1L7, Canada.,Tropical Disease Unit, Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
21
|
Novel morphometric analysis of higher order structure of human radial peri-papillary capillaries: relevance to retinal perfusion efficiency and age. Sci Rep 2019; 9:13464. [PMID: 31530831 PMCID: PMC6748979 DOI: 10.1038/s41598-019-49443-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 08/24/2019] [Indexed: 11/30/2022] Open
Abstract
We apply novel analyses to images of superficial capillaries that are located near and around the optic disc of the human retina: the radial peri-papillary capillaries (RPCs). Due to their unique perfusion of the nerve fibre layer the RPCs are particularly significant for optic-neuropathies. The inputs to the analysis were z-stacks from 3D confocal fluorescence microscopy from 62 human retinas aged 9 to 84 years. Our aim was to find morphometric correlates of age. The retinas had no ophthalmic history. The analysis was undertaken in two stages: (1) converting the z-stacks to 3D tubular networks of vessels, and (2) characterizing the tubular networks using features derived from the Minkowski functionals (MFs). The MFs measure: the capillary volume, surface area, mean breadth, and Euler number. The mean breadth is related to tortuosity, wall shear stress and resistance to flow, and the Euler number is related to the density of loops (collaterals). Features derived from the surface area, mean breadth and Euler number were most related to age (all p ≤ 0.006). The results indicate the importance of pressure-equalizing loops and tortuosity as quantitative measures related to perfusion efficiency. The novel morphometric analysis could quantify disease-related accelerated aging and vessel malformation.
Collapse
|
22
|
De Clercq K, Persoons E, Napso T, Luyten C, Parac-Vogt TN, Sferruzzi-Perri AN, Kerckhofs G, Vriens J. High-resolution contrast-enhanced microCT reveals the true three-dimensional morphology of the murine placenta. Proc Natl Acad Sci U S A 2019; 116:13927-13936. [PMID: 31249139 PMCID: PMC6683600 DOI: 10.1073/pnas.1902688116] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genetic engineering of the mouse genome identified many genes that are essential for embryogenesis. Remarkably, the prevalence of concomitant placental defects in embryonic lethal mutants is highly underestimated and indicates the importance of detailed placental analysis when phenotyping new individual gene knockouts. Here we introduce high-resolution contrast-enhanced microfocus computed tomography (CE-CT) as a nondestructive, high-throughput technique to evaluate the 3D placental morphology. Using a contrast agent, zirconium-substituted Keggin polyoxometalate (Zr-POM), the soft tissue of the placenta (i.e., different layers and cell types and its vasculature) was imaged with a resolution of 3.5 µm voxel size. This approach allowed us to visualize and study early and late stages of placental development. Moreover, CE-CT provides a method to precisely quantify placental parameters (i.e., volumes, volume fraction, ratio of different placental layers, and volumes of specific cell populations) that are crucial for statistical comparison studies. The CE-CT assessment of the 3D morphology of the placentas was validated (i) by comparison with standard histological studies; (ii) by evaluating placentas from 2 different mouse strains, 129S6 and C57BL/6J mice; and (iii) by confirming the placental phenotype of mice lacking phosphoinositol 3-kinase (PI3K)-p110α. Finally, the Zr-POM-based CE-CT allowed for inspection of the vasculature structure in the entire placenta, as well as detecting placental defects in pathologies characterized by embryonic resorption and placental fusion. Taken together, Zr-POM-based CE-CT offers a quantitative 3D methodology to investigate placental development or pathologies.
Collapse
Affiliation(s)
- Katrien De Clercq
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, Gynecology-Pediatrics and Urology Research Group (G-PURE), Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Centre for Brain & Disease Research, 3000 Leuven, Belgium
| | - Eleonora Persoons
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, Gynecology-Pediatrics and Urology Research Group (G-PURE), Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, 3000 Leuven, Belgium
- Vlaams Instituut voor Biotechnologie (VIB) Centre for Brain & Disease Research, 3000 Leuven, Belgium
| | - Tina Napso
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Catherine Luyten
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, Gynecology-Pediatrics and Urology Research Group (G-PURE), Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium
| | - Tatjana N Parac-Vogt
- Molecular Design and Synthesis, Department of Chemistry, KU Leuven, 3000 Leuven, Belgium
| | - Amanda N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | - Greet Kerckhofs
- Biomechanics Laboratory, Institute of Mechanics, Materials, and Civil Engineering, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
- Department of Materials Science and Engineering, KU Leuven, 3000 Leuven, Belgium
- Prometheus, Division of Skeletal Tissue Engineering, KU Leuven, 3000 Leuven, Belgium
| | - Joris Vriens
- Laboratory of Endometrium, Endometriosis & Reproductive Medicine, Department of Development and Regeneration, Gynecology-Pediatrics and Urology Research Group (G-PURE), Katholieke Universiteit (KU) Leuven, 3000 Leuven, Belgium;
| |
Collapse
|
23
|
Adult Pgf -/- mice behaviour and neuroanatomy are altered by neonatal treatment with recombinant placental growth factor. Sci Rep 2019; 9:9285. [PMID: 31243296 PMCID: PMC6594955 DOI: 10.1038/s41598-019-45824-6] [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: 11/19/2018] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Offspring of preeclamptic pregnancies have cognitive alterations. Placental growth factor (PGF), is low in preeclampsia; reduced levels may affect brain development. PGF-null mice differ from normal congenic controls in cerebrovasculature, neuroanatomy and behavior. Using brain imaging and behavioral testing, we asked whether developmentally asynchronous (i.e. neonatal) PGF supplementation alters the vascular, neuroanatomic and/or behavioral status of Pgf−/− mice at adulthood. C57BL/6-Pgf−/− pups were treated intraperitoneally on postnatal days 1–10 with vehicle or PGF at 10 pg/g, 70 pg/g or 700 pg/g. These mice underwent behavioral testing and perfusion for MRI and analysis of retinal vasculature. A second cohort of vehicle- or PGF-treated mice was perfused for micro-CT imaging. 10 pg/g PGF-treated mice exhibited less locomotor activity and greater anxiety-like behavior relative to vehicle-treated mice. Depressive-like behavior showed a sex-specific, dose-dependent decrease and was lowest in 700 pg/g PGF-treated females relative to vehicle-treated females. Spatial learning did not differ. MRI revealed smaller volume of three structures in the 10 pg/g group, larger volume of seven structures in the 70 pg/g group and smaller volume of one structure in the 700 pg/g group. No cerebral or retinal vascular differences were detected. Overall, neonatal PGF replacement altered behavior and neuroanatomy of adult Pgf−/− mice.
Collapse
|
24
|
Kay VR, Rätsep MT, Cahill LS, Hickman AF, Zavan B, Newport ME, Ellegood J, Laliberte CL, Reynolds JN, Carmeliet P, Tayade C, Sled JG, Croy BA. Effects of placental growth factor deficiency on behavior, neuroanatomy, and cerebrovasculature of mice. Physiol Genomics 2018; 50:862-875. [PMID: 30118404 DOI: 10.1152/physiolgenomics.00076.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Preeclampsia, a hypertensive syndrome occurring in 3-5% of human pregnancies, has lifelong health consequences for fetuses. Cognitive ability throughout life is altered, and adult stroke risk is increased. One potential etiological factor for altered brain development is low concentrations of proangiogenic placental growth factor (PGF). Impaired PGF production may promote an antiangiogenic fetal environment during neural and cerebrovascular development. We previously reported delayed vascularization of the hindbrain, altered retinal vascular organization, and less connectivity in the circle of Willis in Pgf-/- mice. We hypothesized Pgf-/- mice would have impaired cognition and altered brain neuroanatomy in addition to compromised cerebrovasculature. Cognitive behavior was assessed in adult Pgf-/- and Pgf+/+ mice by four paradigms followed by postmortem high-resolution MRI of neuroanatomy. X-ray microcomputed tomography imaging investigated the three-dimensional cerebrovascular geometry in another cohort. Pgf-/- mice exhibited poorer spatial memory, less depressive-like behavior, and superior recognition of novel objects. Significantly smaller volumes of 10 structures were detected in the Pgf-/- compared with Pgf+/+ brain. Pgf-/- brain had more total blood vessel segments in the small-diameter range. Lack of PGF altered cognitive functions, brain neuroanatomy, and cerebrovasculature in mice. Pgf-/- mice may be a preclinical model for the offspring effects of low-PGF preeclampsia gestation.
Collapse
Affiliation(s)
- Vanessa R Kay
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Matthew T Rätsep
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | - Andrew F Hickman
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Bruno Zavan
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada.,Federal University of Alfenas (UNIFAL), Alfenas, Minas Gerais , Brazil
| | - Margaret E Newport
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - Jacob Ellegood
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada
| | | | - James N Reynolds
- Centre for Neuroscience Studies, Queen's University , Kingston, Ontario , Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB - Vesalius Research Center, University of Leuven, Department of Oncology , Leuven , Belgium
| | - Chandrakant Tayade
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children , Toronto, Ontario , Canada.,Department of Medical Biophysics, University of Toronto , Ontario , Canada
| | - B Anne Croy
- Department of Biomedical and Molecular Sciences, Queen's University , Kingston, Ontario , Canada
| |
Collapse
|
25
|
HIV antiretroviral exposure in pregnancy induces detrimental placenta vascular changes that are rescued by progesterone supplementation. Sci Rep 2018; 8:6552. [PMID: 29700323 PMCID: PMC5919912 DOI: 10.1038/s41598-018-24680-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 04/03/2018] [Indexed: 11/09/2022] Open
Abstract
Adverse birth outcomes are common in HIV-positive pregnant women receiving combination antiretroviral therapy (cART), especially when cART is initiated in early pregnancy. The mechanisms remain poorly understood. Using a mouse model we demonstrate that protease inhibitor based-cART exposure beginning on day 1 of pregnancy was associated with a pro-angiogenic/pro-branching shift in the placenta driven by lower Flt-1 levels and higher Gcm-1 expression. Micro-CT imaging revealed an increase in the number of arterioles in cART-treated placentas, which correlated with fetal growth restriction. Delaying initiation of cART, or supplementing cART-treated mice with progesterone, prevented the pro-angiogenic/pro-branching shift and the associated placenta vascular changes. In agreement with our mouse findings, we observed an increase in the number of terminal-villi capillaries in placentas from HIV-positive cART-exposed women compared to HIV-negative controls. Capillary number was inversely correlated to maternal progesterone levels. Our study provides evidence that cART exposure during pregnancy influences placenta vascular formation that may in turn contribute to fetal growth restriction. Our findings highlight the need for closer investigation of the placenta in HIV-positive pregnancies, particularly for pregnancies exposed to cART from conception, and suggest that progesterone supplementation could be investigated as a possible intervention to improve placenta function in HIV-positive pregnant women.
Collapse
|
26
|
3D morphological analysis of the mouse cerebral vasculature: Comparison of in vivo and ex vivo methods. PLoS One 2017; 12:e0186676. [PMID: 29053753 PMCID: PMC5650181 DOI: 10.1371/journal.pone.0186676] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 10/05/2017] [Indexed: 02/06/2023] Open
Abstract
Ex vivo 2-photon fluorescence microscopy (2PFM) with optical clearing enables vascular imaging deep into tissue. However, optical clearing may also produce spherical aberrations if the objective lens is not index-matched to the clearing material, while the perfusion, clearing, and fixation procedure may alter vascular morphology. We compared in vivo and ex vivo 2PFM in mice, focusing on apparent differences in microvascular signal and morphology. Following in vivo imaging, the mice (four total) were perfused with a fluorescent gel and their brains fructose-cleared. The brain regions imaged in vivo were imaged ex vivo. Vessels were segmented in both images using an automated tracing algorithm that accounts for the spatially varying PSF in the ex vivo images. This spatial variance is induced by spherical aberrations caused by imaging fructose-cleared tissue with a water-immersion objective. Alignment of the ex vivo image to the in vivo image through a non-linear warping algorithm enabled comparison of apparent vessel diameter, as well as differences in signal. Shrinkage varied as a function of diameter, with capillaries rendered smaller ex vivo by 13%, while penetrating vessels shrunk by 34%. The pial vasculature attenuated in vivo microvascular signal by 40% 300 μm below the tissue surface, but this effect was absent ex vivo. On the whole, ex vivo imaging was found to be valuable for studying deep cortical vasculature.
Collapse
|
27
|
Pratt R, Hutchinson JC, Melbourne A, Zuluaga MA, Virasami A, Vercauteren T, Ourselin S, Sebire NJ, Arthurs OJ, David AL. Imaging the human placental microcirculation with micro-focus computed tomography: Optimisation of tissue preparation and image acquisition. Placenta 2017; 60:36-39. [PMID: 29208237 PMCID: PMC5730539 DOI: 10.1016/j.placenta.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/22/2017] [Accepted: 09/25/2017] [Indexed: 11/18/2022]
Abstract
Micro-CT provides 3D volume imaging with spatial resolution at the micrometre scale. We investigated the optimal human placenta tissue preparation (contrast agent, perfusion pressure, perfusion location and perfusion vessel) and imaging (energy, target material, exposure time and frames) parameters. Microfil (Flow Tech, Carver, MA) produced better fill than Barium sulphate (84.1%(±11.5%)vs70.4%(±18.02%) p = 0.01). Perfusion via umbilical artery produced better fill than via chorionic vessels (83.8%(±17.7%)vs78.0%(±21.9%), p < 0.05), or via umbilical vein (83.8%(±16.4%)vs69.8%(±20.3%), p < 0.01). Imaging at 50 keV with a molybdenum target produced the best contrast to noise ratio. We propose this method to enable quantification and comparison of the human fetoplacental vascular tree. μCT generates high resolution quantifiable placental vascular imaging. We present an optimised perfusion technique for human placental μCT imaging. We present optimised μCT parameters for human placental angiography.
Collapse
Affiliation(s)
- Rosalind Pratt
- Translational Imaging Group, Dept. Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, Gower Street, London WC1E 6BT, UK; Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK.
| | - J Ciaran Hutchinson
- University College London Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK; Department of Histopathology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH London, UK
| | - Andrew Melbourne
- Translational Imaging Group, Dept. Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, Gower Street, London WC1E 6BT, UK
| | - Maria A Zuluaga
- Translational Imaging Group, Dept. Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, Gower Street, London WC1E 6BT, UK
| | - Alex Virasami
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH London, UK
| | - Tom Vercauteren
- Translational Imaging Group, Dept. Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, Gower Street, London WC1E 6BT, UK; Wellcome / EPSRC Centre for Surgical and Interventional Sciences, University College London, London, UK
| | - Sebastien Ourselin
- Translational Imaging Group, Dept. Medical Physics and Biomedical Engineering, University College London, Malet Place Engineering Building, Gower Street, London WC1E 6BT, UK; Wellcome / EPSRC Centre for Surgical and Interventional Sciences, University College London, London, UK
| | - Neil J Sebire
- University College London Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK; Department of Histopathology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH London, UK
| | - Owen J Arthurs
- University College London Great Ormond Street Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK; Paediatric Radiology, Great Ormond Street Hospital for Children NHS Trust, Great Ormond Street, London, WC1N 3JH London, UK
| | - Anna L David
- Institute for Women's Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK; Wellcome / EPSRC Centre for Surgical and Interventional Sciences, University College London, London, UK
| |
Collapse
|
28
|
Rahman A, Cahill LS, Zhou YQ, Hoggarth J, Rennie MY, Seed M, Macgowan CK, Kingdom JC, Adamson SL, Sled JG. A mouse model of antepartum stillbirth. Am J Obstet Gynecol 2017; 217:443.e1-443.e11. [PMID: 28619691 DOI: 10.1016/j.ajog.2017.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/01/2017] [Accepted: 06/06/2017] [Indexed: 11/26/2022]
Abstract
BACKGROUND Many stillbirths of normally formed fetuses in the third trimester could be prevented via delivery if reliable means to anticipate this outcome existed. However, because the etiology of these stillbirths is often unexplained and although the underlying mechanism is presumed to be hypoxia from placental insufficiency, the placentas often appear normal on histopathological examination. Gestational age is a risk factor for antepartum stillbirth, with a rapid rise in stillbirth rates after 40 weeks' gestation. We speculate that a common mechanism may explain antepartum stillbirth in both the late-term and postterm periods. Mice also show increasing rates of stillbirth when pregnancy is artificially prolonged. The model therefore affords an opportunity to characterize events that precede stillbirth. OBJECTIVE The objective of the study was to prolong gestation in mice and monitor fetal and placental growth and cardiovascular changes. STUDY DESIGN From embryonic day 15.5 to embryonic day 18.5, pregnant CD-1 mice received daily progesterone injections to prolong pregnancy by an additional 24 hour period (to embryonic day 19.5). To characterize fetal and placental development, experimental assays were performed throughout late gestation (embryonic day 15.5 to embryonic day 19.5), including postnatal day 1 pups as controls. In addition to collecting fetal and placental weights, we monitored fetal blood flow using Doppler ultrasound and examined the fetoplacental arterial vascular geometry using microcomputed tomography. Evidence of hypoxic organ injury in the fetus was assessed using magnetic resonance imaging and pimonidazole immunohistochemistry. RESULTS At embryonic day 19.5, mean fetal weights were reduced by 14% compared with control postnatal day 1 pups. Ultrasound biomicroscopy showed that fetal heart rate and umbilical artery flow continued to increase at embryonic day 19.5. Despite this, the embryonic day 19.5 fetuses had significant pimonidazole staining in both brain and liver tissue, indicating fetal hypoxia. Placental weights at embryonic day 19.5 were 21% lower than at term (embryonic day 18.5). Microcomputed tomography showed no change in quantitative morphology of the fetoplacental arterial vasculature between embryonic day 18.5 and embryonic day 19.5. CONCLUSION Prolongation of pregnancy renders the murine fetus vulnerable to significant growth restriction and hypoxia because of differential loss of placental mass rather than any compromise in fetoplacental blood flow. Our data are consistent with a hypoxic mechanism of antepartum fetal death in human term and postterm pregnancy and validates the inability of umbilical artery Doppler to safely monitor such fetuses. New tests of placental function are needed to identify the late-term fetus at risk of hypoxia to intervene by delivery to avoid antepartum stillbirth.
Collapse
|
29
|
Cahill LS, Rennie MY, Hoggarth J, Yu LX, Rahman A, Kingdom JC, Seed M, Macgowan CK, Sled JG. Feto- and utero-placental vascular adaptations to chronic maternal hypoxia in the mouse. J Physiol 2017; 596:3285-3297. [PMID: 28861917 DOI: 10.1113/jp274845] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/25/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Chronic fetal hypoxia is one of the most common complications of pregnancy and is known to cause fetal growth restriction. The structural adaptations of the placental vasculature responsible for growth restriction with chronic hypoxia are not well elucidated. Using a mouse model of chronic maternal hypoxia in combination with micro-computed tomography and scanning electron microscopy, we found several placental adaptations that were beneficial to fetal growth including capillary expansion, thinning of the interhaemal membrane and increased radial artery diameters, resulting in a large drop in total utero-placental vascular resistance. One of the mechanisms used to achieve the rapid increase in capillaries was intussusceptive angiogenesis, a strategy used in human placental development to form terminal gas-exchanging villi. These results contribute to our understanding of the structural mechanisms of the placental vasculature responsible for fetal growth restriction and provide a baseline for understanding adaptive physiological responses of the placenta to chronic hypoxia. ABSTRACT The fetus and the placenta in eutherian mammals have a unique set of compensatory mechanisms to respond to several pregnancy complications including chronic maternal hypoxia. This study examined the structural adaptations of the feto- and utero-placental vasculature in an experimental mouse model of chronic maternal hypoxia (11% O2 from embryonic day (E) 14.5-E17.5). While placental weights were unaffected by exposure to chronic hypoxia, using micro-computed tomography, we found a 44% decrease in the absolute feto-placental arterial vascular volume and a 30% decrease in total vessel segments in the chronic hypoxia group compared to control group. Scanning electron microscopy imaging showed significant expansion of the capillary network; consequently, the interhaemal membrane was 11% thinner to facilitate maternal-fetal exchange in the chronic hypoxia placentas. One of the mechanisms for the rapid capillary expansion was intussusceptive angiogenesis. Analysis of the utero-placental arterial tree showed significant increases (24%) in the diameter of the radial arteries, resulting in a decrease in the total utero-placental resistance by 2.6-fold in the mice exposed to chronic maternal hypoxia. Together these adaptations acted to preserve placental weight whereas fetal weight was decreased.
Collapse
Affiliation(s)
- Lindsay S Cahill
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Monique Y Rennie
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Johnathan Hoggarth
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lisa X Yu
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anum Rahman
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - John C Kingdom
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada.,Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Mike Seed
- Division of Cardiology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Christopher K Macgowan
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - John G Sled
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada.,Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Rennie MY, Cahill LS, Adamson SL, Sled JG. Arterio-venous fetoplacental vascular geometry and hemodynamics in the mouse placenta. Placenta 2017; 58:46-51. [PMID: 28962695 DOI: 10.1016/j.placenta.2017.08.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/08/2017] [Accepted: 08/11/2017] [Indexed: 10/19/2022]
Abstract
INTRODUCTION The fetoplacental vasculature network is essential for the exchange of nutrients, gases and wastes with the maternal circulation and for normal fetal development. The present study quantitatively compares arterial and venous morphological and functional differences in the mouse fetoplacental vascular network. METHODS High resolution X-ray micro-computed tomography was used to visualize the 3D geometry of the arterial and venous fetoplacental vasculature in embryonic day 15.5 CD-1 mice (n = 5). Automated image analysis was used to measure the vascular geometry of the approximately 4100 arterial segments and 3200 venous segments per specimen to simulate blood flow through these networks. RESULTS Both the arterial and venous trees demonstrated a hierarchical branching structure with 8 or 9 (arterial) or 8 (venous) orders. The venous tree was smaller in volume and overall dimensions than the arterial tree. Venous vessel diameters increased more rapidly than arteries with each successive order, leading to lower overall resistance, although the umbilical vein was notably smaller and of higher resistance than these scaling relationships would predict. Simulation of blood flow for these vascular networks showed that 57% of total resistance resides in the umbilical artery and arterial tree, 17% in the capillary bed, and 26% in the venous tree and umbilical vein. DISCUSSION A detailed examination of the mouse fetoplacental arterial and venous tree revealed features, such as the distribution of resistance and the dimension of the venous tree, that were both morphologically distinct from other vascular beds and that appeared adapted to the specialized requirements of sustaining a fetus.
Collapse
Affiliation(s)
- Monique Y Rennie
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Lindsay S Cahill
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - S Lee Adamson
- Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario Canada
| | - John G Sled
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Translational Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
31
|
Activated NK cells cause placental dysfunction and miscarriages in fetal alloimmune thrombocytopenia. Nat Commun 2017; 8:224. [PMID: 28794456 PMCID: PMC5550461 DOI: 10.1038/s41467-017-00269-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 06/16/2017] [Indexed: 01/29/2023] Open
Abstract
Miscarriage and intrauterine growth restriction (IUGR) are devastating complications in fetal/neonatal alloimmune thrombocytopenia (FNAIT). We previously reported the mechanisms for bleeding diatheses, but it is unknown whether placental, decidual immune cells or other abnormalities at the maternal-fetal interface contribute to FNAIT. Here we show that maternal immune responses to fetal platelet antigens cause miscarriage and IUGR that are associated with vascular and immune pathologies in murine FNAIT models. Uterine natural killer (uNK) cell recruitment and survival beyond mid-gestation lead to elevated NKp46 and CD107 expression, perforin release and trophoblast apoptosis. Depletion of NK cells restores normal spiral artery remodeling and placental function, prevents miscarriage, and rescues hemorrhage in neonates. Blockade of NK activation receptors (NKp46, FcɣRIIIa) also rescues pregnancy loss. These findings shed light on uNK antibody-dependent cell-mediated cytotoxicity of invasive trophoblasts as a pathological mechanism in FNAIT, and suggest that anti-NK cell therapies may prevent immune-mediated pregnancy loss and ameliorate FNAIT.Fetal/neonatal alloimmune thrombocytopenia (FNAIT) is a gestational disease caused by maternal immune responses against fetal platelets. Using a FNAIT mouse model and human trophoblast cell lines, here the authors show that uterine natural killer cell-mediated trophoblast apoptosis contributes to FNAIT pathogenesis.
Collapse
|
32
|
Chen B, Duan J, Chabot-Lecoanet AC, Lu H, Tonnelet R, Morel O, Beaumont M. Ex vivo magnetic resonance angiography to explore placental vascular anatomy. Placenta 2017; 58:40-45. [PMID: 28962694 DOI: 10.1016/j.placenta.2017.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/23/2017] [Accepted: 08/01/2017] [Indexed: 11/19/2022]
Abstract
INTRODUCTION A normal placenta development is crucial for a successful pregnancy. In case of major obstetric complications such as intra-uterine growth restriction, the placental vascularization morphological alteration at macroscopic level is less known than that at microscopic scale. Ex vivo MRA has the potential to visualize whole fresh human placental vasculature fast and efficiently but can be hampered by contrast agent extravasation problem. This study aimed to provide an optimized ex vivo MRA protocol to acquire understanding of global human placenta vasculature morphology. METHODS Six fresh normal human placentas were imaged with two contrast agents (i.e. Gd-chelate and pump oil) using different imaging parameters on a 3T clinical MR scanner (GE). The contrast to noise ratio, signal to noise ratio and enhancement efficiency were assessed in order to decide which contrast agent and imaging protocol was better. In the end, morphology indices were measured based on the 3D vasculature models reconstructed from the placentas imaged with the optimized protocol. RESULTS With the same imaging parameters, the CNR and the enhancement efficiency of images enhanced with pump oil were superior to those using Gd-chelate. Enhanced by pump oil, an optimized ex vivo MRA protocol was determined, leading to a clear 3D visualization and reconstruction of human placenta vascularization. DISCUSSION The proposed ex vivo MRA method is easy to manipulate, and can be used to investigate the human placental vasculature morphology. The acquired data are of good quality and can be used for characterization of placenta vascularization morphology.
Collapse
Affiliation(s)
- Bailiang Chen
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France; CIC-IT Nancy, Inserm CIC1433, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France
| | - Jie Duan
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France; Regional Maternity of University of Nancy, 10 Rue Heydenreich, 54000, Nancy, France
| | - Anne-Claire Chabot-Lecoanet
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France; Regional Maternity of University of Nancy, 10 Rue Heydenreich, 54000, Nancy, France
| | - Huanrong Lu
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France
| | - Romain Tonnelet
- The Neurointerventional Department, CHRU of Nancy, 54000, Nancy, France
| | - Oliver Morel
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France; Regional Maternity of University of Nancy, 10 Rue Heydenreich, 54000, Nancy, France
| | - Marine Beaumont
- IADI Inserm U947, Lorraine University, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France; CIC-IT Nancy, Inserm CIC1433, 4 Rue Du Morvan, 54500, Vandoeuvre lès Nancy, France.
| |
Collapse
|
33
|
Viscosity and haemodynamics in a late gestation rat feto-placental arterial network. Biomech Model Mechanobiol 2017; 16:1361-1372. [PMID: 28258413 DOI: 10.1007/s10237-017-0892-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/21/2017] [Indexed: 12/26/2022]
Abstract
The placenta is a transient organ which develops during pregnancy to provide haemotrophic support for healthy fetal growth and development. Fundamental to its function is the healthy development of vascular trees in the feto-placental arterial network. Despite the strong association of haemodynamics with vascular remodelling mechanisms, there is a lack of computational haemodynamic data that may improve our understanding of feto-placental physiology. The aim of this work was to create a comprehensive 3D computational fluid dynamics model of a substructure of the rat feto-placental arterial network and investigate the influence of viscosity on wall shear stress (WSS). Late gestation rat feto-placental arteries were perfused with radiopaque Microfil and scanned via micro-computed tomography to capture the feto-placental arterial geometry in 3D. A detailed description of rat fetal blood viscosity parameters was developed, and three different approaches to feto-placental haemodynamics were simulated in 3D using the finite volume method: Newtonian model, non-Newtonian Carreau-Yasuda model and Fåhræus-Lindqvist effect model. Significant variability in WSS was observed between different viscosity models. The physiologically-realistic simulations using the Fåhræus-Lindqvist effect and rat fetal blood estimates of viscosity revealed detailed patterns of WSS throughout the arterial network. We found WSS gradients at bifurcation regions, which may contribute to vessel enlargement, and sprouting and pruning during angiogenesis. This simulation of feto-placental haemodynamics shows the heterogeneous WSS distribution throughout the network and demonstrates the ability to determine physiologically-relevant WSS magnitudes, patterns and gradients. This model will help advance our understanding of vascular physiology and remodelling in the feto-placental network.
Collapse
|
34
|
Dorr A, Thomason LA, Koletar MM, Joo IL, Steinman J, Cahill LS, Sled JG, Stefanovic B. Effects of voluntary exercise on structure and function of cortical microvasculature. J Cereb Blood Flow Metab 2017; 37:1046-1059. [PMID: 27683451 PMCID: PMC5363487 DOI: 10.1177/0271678x16669514] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Aerobic activity has been shown highly beneficial to brain health, yet much uncertainty still surrounds the effects of exercise on the functioning of cerebral microvasculature. This study used two-photon fluorescence microscopy to examine cerebral hemodynamic alterations as well as accompanying geometric changes in the cortical microvascular network following five weeks of voluntary exercise in transgenic mice endogenously expressing tdTomato in vascular endothelial cells to allow visualization of microvessels irrespective of their perfusion levels. We found a diminished microvascular response to a hypercapnic challenge (10% FiCO2) in running mice when compared to that in nonrunning controls despite commensurate increases in transcutaneous CO2 tension. The flow increase to hypercapnia in runners was 70% lower than that in nonrunners (p = 0.0070) and the runners' arteriolar red blood cell speed changed by only half the amount seen in nonrunners (p = 0.0085). No changes were seen in resting hemodynamics or in the systemic physiological parameters measured. Although a few unperfused new vessels were observed on visual inspection, running did not produce significant morphological differences in the microvascular morphometric parameters, quantified following semiautomated tracking of the microvascular networks. We propose that voluntary running led to increased cortical microvascular efficiency and desensitization to CO2 elevation.
Collapse
Affiliation(s)
| | | | | | - Illsung L Joo
- 1 Sunnybrook Research Institute, Toronto, Canada.,2 Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Joe Steinman
- 2 Department of Medical Biophysics, University of Toronto, Toronto, Canada.,3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - Lindsay S Cahill
- 3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - John G Sled
- 2 Department of Medical Biophysics, University of Toronto, Toronto, Canada.,3 Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Canada
| | - Bojana Stefanovic
- 1 Sunnybrook Research Institute, Toronto, Canada.,2 Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
35
|
Chi L, Ahmed A, Roy AR, Vuong S, Cahill LS, Caporiccio L, Sled JG, Caniggia I, Wilson MD, Delgado-Olguin P. Ehmt2/G9a controls placental vascular maturation by activating the Notch pathway. Development 2017; 144:1976-1987. [DOI: 10.1242/dev.148916] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/13/2017] [Indexed: 12/20/2022]
Abstract
Defective fetoplacental vascular maturation causes intrauterine growth restriction (IUGR). A transcriptional switch initiates placental maturation where blood vessels elongate. However, cellular mechanisms and regulatory pathways involved are unknown. We show that the histone methyltransferase Ehmt2, also known as G9a, activates the Notch pathway to promote placental vascular maturation. Placental vasculature from embryos with G9a-deficient endothelial progenitor cells failed to expand due to decreased endothelial cell proliferation and increased trophoblast proliferation. Moreover, G9a deficiency altered the transcriptional switch initiating placental maturation and caused downregulation of Notch pathway effectors including Rbpj. Importantly, Notch pathway activation in G9a-deficient endothelial progenitors extended embryonic life and rescued placental vascular expansion. Thus, G9a activates the Notch pathway to balance endothelial cell and trophoblast proliferation and coordinates the transcriptional switch controlling placental vascular maturation. Accordingly, G9A and RBPJ were downregulated in human placentae from IUGR-affected pregnancies, suggesting that G9a is an important regulator in placental diseases caused by defective vascular maturation.
Collapse
Affiliation(s)
- Lijun Chi
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
| | - Abdalla Ahmed
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Anna R. Roy
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sandra Vuong
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Lindsay S. Cahill
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Laura Caporiccio
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
| | - John G. Sled
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Isabella Caniggia
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Michael D. Wilson
- Program in Genetics & Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Paul Delgado-Olguin
- Translational Medicine, The Hospital for Sick Children, Toronto, ON, M5G0A4, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, ON, Canada
| |
Collapse
|
36
|
Gagnon L, Smith AF, Boas DA, Devor A, Secomb TW, Sakadžić S. Modeling of Cerebral Oxygen Transport Based on In vivo Microscopic Imaging of Microvascular Network Structure, Blood Flow, and Oxygenation. Front Comput Neurosci 2016; 10:82. [PMID: 27630556 PMCID: PMC5006088 DOI: 10.3389/fncom.2016.00082] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 07/25/2016] [Indexed: 01/09/2023] Open
Abstract
Oxygen is delivered to brain tissue by a dense network of microvessels, which actively control cerebral blood flow (CBF) through vasodilation and contraction in response to changing levels of neural activity. Understanding these network-level processes is immediately relevant for (1) interpretation of functional Magnetic Resonance Imaging (fMRI) signals, and (2) investigation of neurological diseases in which a deterioration of neurovascular and neuro-metabolic physiology contributes to motor and cognitive decline. Experimental data on the structure, flow and oxygen levels of microvascular networks are needed, together with theoretical methods to integrate this information and predict physiologically relevant properties that are not directly measurable. Recent progress in optical imaging technologies for high-resolution in vivo measurement of the cerebral microvascular architecture, blood flow, and oxygenation enables construction of detailed computational models of cerebral hemodynamics and oxygen transport based on realistic three-dimensional microvascular networks. In this article, we review state-of-the-art optical microscopy technologies for quantitative in vivo imaging of cerebral microvascular structure, blood flow and oxygenation, and theoretical methods that utilize such data to generate spatially resolved models for blood flow and oxygen transport. These “bottom-up” models are essential for the understanding of the processes governing brain oxygenation in normal and disease states and for eventual translation of the lessons learned from animal studies to humans.
Collapse
Affiliation(s)
- Louis Gagnon
- Optics Division, Department of Radiology, MHG/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Amy F Smith
- Institut de Mécanique des Fluides de ToulouseToulouse, France; Department of Physiology, University of ArizonaTucson, AZ, USA
| | - David A Boas
- Optics Division, Department of Radiology, MHG/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| | - Anna Devor
- Optics Division, Department of Radiology, MHG/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical SchoolCharlestown, MA, USA; Departments of Neurosciences and Radiology, University of California, San DiegoLa Jolla, CA, USA
| | | | - Sava Sakadžić
- Optics Division, Department of Radiology, MHG/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School Charlestown, MA, USA
| |
Collapse
|
37
|
Palatnik A, Xin H, Su EJ. Dichotomous effects of aryl hydrocarbon receptor (AHR) activation on human fetoplacental endothelial cell function. Placenta 2016; 44:61-8. [PMID: 27452439 DOI: 10.1016/j.placenta.2016.06.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/26/2016] [Accepted: 06/10/2016] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Maternal cigarette smoking is associated with elevated fetoplacental vascular resistance and fetal growth restriction (FGR). While studies have demonstrated varying effects of nicotine on blood flow, the role of polycyclic aromatic hydrocarbons (PAHs), abundant toxins in cigarette smoke that cross the placenta, has not been elucidated. We hypothesized that exposure of human fetoplacental endothelial cells (ECs) to the PAH benzo[a]yrene (BaP) would result in up-regulation of cyclooxygenase-2 (PTGS2) and preferential production of vasoconstrictive prostanoids via activation of the aryl hydrocarbon receptor (AHR) pathway. METHODS ECs were isolated, cultured, and treated with vehicle or BaP. ECs were subjected to real-time PCR, western blotting, enzyme immunoassays, wound scratch assays, tube formation assays, and RNA interference against AHR. Statistical analyses were performed with Student's t-test, one-way ANOVA followed by multiple comparisons testing when appropriate, or the Kruskal-Wallis H test. RESULTS BaP induced PTGS2 expression (p < 0.05) and production of the stable metabolite of prostacyclin (p = 0.001) in fetoplacental ECs without affecting thromboxane. These effects were ablated by PTGS2 inhibition (p < 0.01) and RNA interference of AHR (p < 0.001). Surprisingly, despite the induction of prostacyclin, EC migration (p = 0.007) and tube formation (p = 0.003) were inhibited by BaP. AHR inhibition, however, rescued tube formation (p = 0.008). DISCUSSION BaP-mediated AHR activation results in induction of PTGS2 expression and enhanced production of prostacyclin metabolite. Despite an increase in this vasodilatory and pro-angiogenic prostanoid, BaP exposure also impairs EC migration and angiogenesis through AHR. This suggests that PAH may adversely affect the fetoplacental vasculature through its regulation of angiogenesis.
Collapse
Affiliation(s)
- Anna Palatnik
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Hong Xin
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Emily J Su
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
38
|
Farmer SA, Nelin TD, Falvo MJ, Wold LE. Ambient and household air pollution: complex triggers of disease. Am J Physiol Heart Circ Physiol 2015; 307:H467-76. [PMID: 24929855 DOI: 10.1152/ajpheart.00235.2014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Concentrations of outdoor air pollution are on the rise, particularly due to rapid urbanization worldwide. Alternatively, poor ventilation, cigarette smoke, and other toxic chemicals contribute to rising concentrations of indoor air pollution. The World Health Organization recently reported that deaths attributable to indoor and outdoor air pollutant exposure are more than double what was originally documented. Epidemiological, clinical, and animal data have demonstrated a clear connection between rising concentrations of air pollution (both indoor and outdoor) and a host of adverse health effects. During the past five years, animal, clinical, and epidemiological studies have explored the adverse health effects associated with exposure to both indoor and outdoor air pollutants throughout the various stages of life. This review provides a summary of the detrimental effects of air pollution through examination of current animal, clinical, and epidemiological studies and exposure during three different periods: maternal (in utero), early life, and adulthood. Additionally, we recommend future lines of research while suggesting conceivable strategies to curb exposure to indoor and outdoor air pollutants.
Collapse
|
39
|
Rennie MY, Rahman A, Whiteley KJ, Sled JG, Adamson SL. Site-specific increases in utero- and fetoplacental arterial vascular resistance in eNOS-deficient mice due to impaired arterial enlargement. Biol Reprod 2014; 92:48. [PMID: 25519187 DOI: 10.1095/biolreprod.114.123968] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The sites of elevated vascular resistance that impede placental perfusion in pathological pregnancies are unknown. In the current study, we identified these sites in a knockout mouse model (eNOS(-/-)) with reduced uterine (-55%) and umbilical (-29%) artery blood flows caused by endothelial nitric oxide synthase deficiency. Uteroplacental and fetoplacental arterial vascular trees of pregnant mice near term were imaged using x-ray microcomputed tomography (n = 5-10 placentas from 3-5 dams/group). The resulting three-dimensional images were analyzed to assess vessel geometry and vascular resistance. In control and eNOS(-/-) trees, ∼90% of total uteroplacental vascular resistance was located in the radial arteries. Changes in eNOS(-/-) vessel geometry, including 30% reductions in uterine, radial, and spiral artery diameters, were calculated to increase arterial resistance downstream of the uterine artery by 2.3-fold, predicting a 57% decrease in uterine blood flow. Despite large reductions in eNOS(-/-) spiral arteries (-55% by volume) and maternal canals (-67% by volume), these vessels were relatively minor contributors to resistance. In the eNOS(-/-) fetoplacental tree, the number of arterioles (50-75 μm diameter) increased by 26%. Nevertheless, calculated resistance rose by 19%, predominantly because arteries near the periphery of the tree selectively exhibited a 7%-9% diameter reduction. We conclude that previously observed decreases in uterine and umbilical blood flows in eNOS(-/-) pregnancies are associated with markedly divergent structural changes in the uteroplacental versus fetoplacental circulations. Results showed the radial arteries were critical determinants of uteroplacental resistance in mice and therefore warrant greater attention in future studies in pathological human pregnancies.
Collapse
Affiliation(s)
- Monique Y Rennie
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Anum Rahman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Kathie J Whiteley
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - John G Sled
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Ontario, Canada Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - S Lee Adamson
- Department of Obstetrics and Gynecology, University of Toronto, Toronto, Ontario, Canada Department of Physiology, University of Toronto, Toronto, Ontario, Canada Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
40
|
Langlois PH, Hoyt AT, Desrosiers TA, Lupo PJ, Lawson CC, Waters MA, Rocheleau CM, Shaw GM, Romitti PA, Gilboa SM, Malik S. Maternal occupational exposure to polycyclic aromatic hydrocarbons and small for gestational age offspring. Occup Environ Med 2014; 71:529-35. [PMID: 24893704 PMCID: PMC4497781 DOI: 10.1136/oemed-2013-101833] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVES While some of the highest maternal exposures to polycyclic aromatic hydrocarbons (PAHs) occur in the workplace, there is only one previous study of occupational PAH exposure and adverse pregnancy outcomes. We sought to extend this literature using interview data combined with detailed exposure assessment. METHODS Data for 1997-2002 were analysed from mothers of infants without major birth defects in the National Birth Defects Prevention Study, a large population-based case-control study in the USA. Maternal telephone interviews yielded information on jobs held in the month before conception through delivery. From 6252 eligible control mothers, 2803 completed the interview, had a job, met other selection criteria, and were included in the analysis. Two industrial hygienists independently assessed occupational exposure to PAHs from the interview and reviewed results with a third to reach consensus. Small for gestational age (SGA) was the only adverse pregnancy outcome with enough exposed cases to yield meaningful results. Logistic regression estimated crude and adjusted ORs. RESULTS Of the 2803 mothers, 221 (7.9%) had infants who were SGA. Occupational PAH exposure was found for 17 (7.7%) of the mothers with SGA offspring and 102 (4.0%) of the remaining mothers. Almost half the jobs with exposure were related to food preparation and serving. After adjustment for maternal age, there was a significant association of occupational exposure with SGA (OR=2.2, 95% CI 1.3 to 3.8). CONCLUSIONS Maternal occupational exposure to PAHs was found to be associated with increased risk of SGA offspring.
Collapse
Affiliation(s)
- Peter H Langlois
- Texas Center for Birth Defects Research and Prevention, Birth Defects Epidemiology and Surveillance Branch, Texas Department of State Health Services, Austin, Texas, USA
| | - Adrienne T Hoyt
- Texas Center for Birth Defects Research and Prevention, Birth Defects Epidemiology and Surveillance Branch, Texas Department of State Health Services, Austin, Texas, USA
| | - Tania A Desrosiers
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Philip J Lupo
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Christina C Lawson
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Cincinnati, Ohio, USA
| | - Martha A Waters
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Cincinnati, Ohio, USA
| | - Carissa M Rocheleau
- National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Cincinnati, Ohio, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, The University of Iowa, Iowa City, Iowa, USA
| | - Suzanne M Gilboa
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Sadia Malik
- Department of Pediatrics, University of Arkansas Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
41
|
Grazul-Bilska AT, Johnson ML, Borowicz PP, Bilski JJ, Cymbaluk T, Norberg S, Redmer DA, Reynolds LP. Placental development during early pregnancy in sheep: effects of embryo origin on vascularization. Reproduction 2014; 147:639-48. [PMID: 24472816 DOI: 10.1530/rep-13-0663] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Utero-placental growth and vascular development are critical for pregnancy establishment that may be altered by various factors including assisted reproductive technologies (ART), nutrition, or others, leading to compromised pregnancy. We hypothesized that placental vascularization and expression of angiogenic factors are altered early in pregnancies after transfer of embryos created using selected ART methods. Pregnancies were achieved through natural mating (NAT), or transfer of embryos from NAT (NAT-ET), or IVF or in vitro activation (IVA). Placental tissues were collected on day 22 of pregnancy. In maternal caruncles (CAR), vascular cell proliferation was less (P<0.05) for IVA than other groups. Compared with NAT, density of blood vessels was less (P<0.05) for IVF and IVA in fetal membranes (FM) and for NAT-ET, IVF, and IVA in CAR. In FM, mRNA expression was decreased (P<0.01-0.08) in NAT-ET, IVF, and IVA compared with NAT for vascular endothelial growth factor (VEGF) and its receptor FLT1, placental growth factor (PGF), neuropilin 1 (NP1) and NP2, angiopoietin 1 (ANGPT1) and ANGPT2, endothelial nitric oxide synthase 3 (NOS3), hypoxia-inducible factor 1A (HIF1A), fibroblast growth factor 2 (FGF2), and its receptor FGFR2. In CAR, mRNA expression was decreased (P<0.01-0.05) in NAT-ET, IVF, and IVA compared with NAT for VEGF, FLT1, PGF, ANGPT1, and TEK. Decreased mRNA expression for 12 of 14 angiogenic factors across FM and CAR in NAT-ET, IVF, and IVA pregnancies was associated with reduced placental vascular development, which would lead to poor placental function and compromised fetal and placental growth and development.
Collapse
Affiliation(s)
- Anna T Grazul-Bilska
- Department of Animal Sciences, Center for Nutrition and Pregnancy, North Dakota State University, Fargo, North Dakota 58108, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ghanavati S, Lerch JP, Sled JG. Automatic anatomical labeling of the complete cerebral vasculature in mouse models. Neuroimage 2014; 95:117-28. [PMID: 24680868 DOI: 10.1016/j.neuroimage.2014.03.044] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 02/14/2014] [Accepted: 03/15/2014] [Indexed: 01/08/2023] Open
Abstract
Study of cerebral vascular structure broadens our understanding of underlying variations, such as pathologies that can lead to cerebrovascular disorders. The development of high resolution 3D imaging modalities has provided us with the raw material to study the blood vessels in small animals such as mice. However, the high complexity and 3D nature of the cerebral vasculature make comparison and analysis of the vessels difficult, time-consuming and laborious. Here we present a framework for automated segmentation and recognition of the cerebral vessels in high resolution 3D images that addresses this need. The vasculature is segmented by following vessel center lines starting from automatically generated seeds and the vascular structure is represented as a graph. Each vessel segment is represented as an edge in the graph and has local features such as length, diameter, and direction, and relational features representing the connectivity of the vessel segments. Using these features, each edge in the graph is automatically labeled with its anatomical name using a stochastic relaxation algorithm. We have validated our method on micro-CT images of C57Bl/6J mice. A leave-one-out test performed on the labeled data set demonstrated the recognition rate for all vessels including major named vessels and their minor branches to be >75%. This automatic segmentation and recognition methods facilitate the comparison of blood vessels in large populations of subjects and allow us to study cerebrovascular variations.
Collapse
Affiliation(s)
- Sahar Ghanavati
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada; Mouse Imaging Centre, The Hospital for Sick Children, 25 Orde St., Toronto, Ontario M5T 3H7, Canada.
| | - Jason P Lerch
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada; Mouse Imaging Centre, The Hospital for Sick Children, 25 Orde St., Toronto, Ontario M5T 3H7, Canada
| | - John G Sled
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5G 2M9, Canada; Mouse Imaging Centre, The Hospital for Sick Children, 25 Orde St., Toronto, Ontario M5T 3H7, Canada
| |
Collapse
|
43
|
Le Vee M, Kolasa E, Jouan E, Collet N, Fardel O. Differentiation of human placental BeWo cells by the environmental contaminant benzo(a)pyrene. Chem Biol Interact 2014; 210:1-11. [DOI: 10.1016/j.cbi.2013.12.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/22/2013] [Accepted: 12/09/2013] [Indexed: 12/15/2022]
|
44
|
Wise LD, Winkelmann CT, Dogdas B, Bagchi A. Micro-computed tomography imaging and analysis in developmental biology and toxicology. ACTA ACUST UNITED AC 2014; 99:71-82. [PMID: 23897592 DOI: 10.1002/bdrc.21033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 05/07/2013] [Indexed: 01/02/2023]
Abstract
Micro-computed tomography (micro-CT) is a high resolution imaging technique that has expanded and strengthened in use since it was last reviewed in this journal in 2004. The technology has expanded to include more detailed analysis of bone, as well as soft tissues, by use of various contrast agents. It is increasingly applied to questions in developmental biology and developmental toxicology. Relatively high-throughput protocols now provide a powerful and efficient means to evaluate embryos and fetuses subjected to genetic manipulations or chemical exposures. This review provides an overview of the technology, including scanning, reconstruction, visualization, segmentation, and analysis of micro-CT generated images. This is followed by a review of more recent applications of the technology in some common laboratory species that highlight the diverse issues that can be addressed.
Collapse
Affiliation(s)
- L David Wise
- Merck Research Laboratories, Departments of Safety Assessment and Laboratory Animal Resources, Imaging, and Informatics IT, West Point, PA, USA.
| | | | | | | |
Collapse
|
45
|
Lubinsky M. A vascular and thrombotic model of gastroschisis. Am J Med Genet A 2014; 164A:915-7. [PMID: 24458365 DOI: 10.1002/ajmg.a.36370] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 10/17/2013] [Indexed: 01/09/2023]
Abstract
A binary vascular/thrombotic pathogenesis for gastroschisis, a form of congenital bowel herniation, is proposed, where normal right umbilical vein involution creates a possible site for thrombosis adjacent to the umbilical ring. If thrombosis occurs, it weakens the area, explaining overwhelmingly right-sided lesions. The model arises from the existence of two groups of risk factors with different maternal age associations. Older mothers show a greater association with vascular factors (although this may actually represent a lack of any significant maternal age effect), consistent with associations of gastroschisis with congenital heart lesions and with amyoplasia. Alternatively, other predispositions, and especially decreased maternal age, the greatest known risk factor, associate with factors raising maternal estrogen, with evidence that estrogen in turn acts here as a predisposition to thrombosis. Absorption of thrombotic by-products from the amniotic fluid can explain the unusual amniocyte inclusions that are common with gastroschisis, while a role for estrogens suggests a connection between rising gastroschisis prevalence and increasing environmental contamination with estrogen disruptors. This model explains a variety of structural and epidemiological findings, and suggests that stratification of data based on binary effects may clarify associated risks and mechanisms. The model also shows that what is often referred to as vascular disruption may actually reflect alternative or additional factors instead, including thrombosis as a primary mechanism.
Collapse
|
46
|
Debbaut C, Segers P, Cornillie P, Casteleyn C, Dierick M, Laleman W, Monbaliu D. Analyzing the human liver vascular architecture by combining vascular corrosion casting and micro-CT scanning: a feasibility study. J Anat 2014; 224:509-17. [PMID: 24433401 DOI: 10.1111/joa.12156] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2013] [Indexed: 11/26/2022] Open
Abstract
Although a full understanding of the hepatic circulation is one of the keys to successfully perform liver surgery and to elucidate liver pathology, relatively little is known about the functional organization of the liver vasculature. Therefore, we materialized and visualized the human hepatic vasculature at different scales, and performed a morphological analysis by combining vascular corrosion casting with novel micro-computer tomography (CT) and image analysis techniques. A human liver vascular corrosion cast was obtained by simultaneous resin injection in the hepatic artery (HA) and portal vein (PV). A high resolution (110 μm) micro-CT scan of the total cast allowed gathering detailed macrovascular data. Subsequently, a mesocirculation sample (starting at generation 5; 88 × 68 × 80 mm³) and a microcirculation sample (terminal vessels including sinusoids; 2.0 × 1.5 × 1.7 mm³) were dissected and imaged at a 71-μm and 2.6-μm resolution, respectively. Segmentations and 3D reconstructions allowed quantifying the macro- and mesoscale branching topology, and geometrical features of HA, PV and hepatic venous trees up to 13 generations (radii ranging from 13.2 mm to 80 μm; lengths from 74.4 mm to 0.74 mm), as well as microvascular characteristics (mean sinusoidal radius of 6.63 μm). Combining corrosion casting and micro-CT imaging allows quantifying the branching topology and geometrical features of hepatic trees using a multiscale approach from the macro- down to the microcirculation. This may lead to novel insights into liver circulation, such as internal blood flow distributions and anatomical consequences of pathologies (e.g. cirrhosis).
Collapse
Affiliation(s)
- Charlotte Debbaut
- Biofluid, Tissue and Solid Mechanics for Medical Applications, Institute Biomedical Technology, Department of Electronics and Information Systems, iMinds Future Health Department, Ghent University, Gent, Belgium
| | | | | | | | | | | | | |
Collapse
|
47
|
Rennie MY, Sled JG, Adamson SL. Effects of Genes and Environment on the Fetoplacental Arterial Microcirculation in Mice Revealed by Micro-Computed Tomography Imaging. Microcirculation 2014; 21:48-57. [DOI: 10.1111/micc.12073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 06/22/2013] [Indexed: 01/24/2023]
Affiliation(s)
- Monique Y. Rennie
- Heart Research Center; Oregon Health and Science University; Portland Oregon USA
| | - John G. Sled
- Mouse Imaging Centre of the Hospital for Sick Children, Department of Medical Biophysics; University of Toronto; Toronto Ontario Canada
| | - S. Lee Adamson
- Samuel Lunenfeld Research Institute of Mount Sinai Hospital; Departments of Obstetrics and Gynaecology, and Physiology; University of Toronto; Toronto Ontario Canada
| |
Collapse
|
48
|
A perfusion procedure for imaging of the mouse cerebral vasculature by X-ray micro-CT. J Neurosci Methods 2013; 221:70-7. [PMID: 24056228 DOI: 10.1016/j.jneumeth.2013.09.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 08/26/2013] [Accepted: 09/02/2013] [Indexed: 12/19/2022]
Abstract
BACKGROUND Micro-CT is a novel X-ray imaging modality which can provide 3D high resolution images of the vascular network filled with contrast agent. The cerebrovascular system is a complex anatomical structure that can be imaged with contrast enhanced micro-CT. However, the morphology of the cerebrovasculature and many circulatory anastomosis in the brain result in high variations in the extent of contrast agent filling in the blood vessels and as a result, the vasculature of different subjects appear differently in the acquired images. Specifically, the posterior circulation is not consistently perfused with the contrast agent in many brain specimens and thus, many major vessels that perfuse blood to the midbrain and hindbrain are not visible in the micro-CT images acquired from these samples. NEW METHOD In this paper, we present a modified surgical procedure of cerebral vasculature perfusion through the left ventricle with Microfil contrast agent, in order to achieve a more uniform perfusion of blood vessels throughout the brain and as a result, more consistent images of the cerebrovasculature. Our method consists of filling the posterior cerebral circulation with contrast agent, followed by the perfusion of the whole cerebrovasculature. RESULTS Our histological results show that over 90% of the vessels in the entire brain, including the cerebellum, were filled with contrast agent. COMPARISON WITH EXISTING METHOD Our results show that the new technique of sample perfusion decreases the variability of the posterior circulation in the cerebellum in micro-CT images by 6.9%. CONCLUSIONS This new technique of sample preparation improves the quality of cerebrovascular images.
Collapse
|
49
|
Venditti CC, Casselman R, Murphy MSQ, Adamson SL, Sled JG, Smith GN. Chronic carbon monoxide inhalation during pregnancy augments uterine artery blood flow and uteroplacental vascular growth in mice. Am J Physiol Regul Integr Comp Physiol 2013; 305:R939-48. [PMID: 23986360 DOI: 10.1152/ajpregu.00204.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
End-tidal breath carbon monoxide (CO) is abnormally low in women with preeclampsia (PE), while women smoking during pregnancy have shown an increase in CO levels and a 33% lower incidence of PE. This effect may be, in part, due to lowered sFLT1 plasma levels in smokers, and perhaps low-level CO inhalation can attenuate the development of PE in high-risk women. Our previous work showed maternal chronic CO exposure (<300 ppm) throughout gestation had no maternal or fetal deleterious effects in mice. Our current study evaluated the uteroplacental vascular effects in CD-1 maternal mice that inhaled CO (250 ppm) both chronically, gestation day (GD) 0.5 to 18.5, and acutely, 2.5 h on each of GD 10.5 and 14.5. We demonstrated, using microultrasound measurements of blood velocity and microcomputed tomography imaging of the uteroplacental vasculature, that chronic maternal exposure to CO doubled uterine artery blood flow and augmented uteroplacental vascular diameters and branching. This finding may be of benefit to women with PE, as they exhibit uteroplacental vascular compromise. The ratio of VEGF protein to its FLT1 receptor was increased in the placenta, suggesting a shift to a more angiogenic state; however, maternal circulating levels of VEGF, sFLT1, and their ratio were not significantly changed. Doppler blood velocities in the maternal uterine artery and fetal umbilical artery and vein were unaltered. This study provides in vivo evidence that chronic inhalation of 250 ppm CO throughout gestation augments uterine blood flow and uteroplacental vascular growth, changes that may protect against the subsequent development of preeclampsia.
Collapse
Affiliation(s)
- Carolina C Venditti
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston General Hospital, Kingston, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Conroy AL, Silver KL, Zhong K, Rennie M, Ward P, Sarma JV, Molyneux ME, Sled J, Fletcher JF, Rogerson S, Kain KC. Complement activation and the resulting placental vascular insufficiency drives fetal growth restriction associated with placental malaria. Cell Host Microbe 2013; 13:215-26. [PMID: 23414761 DOI: 10.1016/j.chom.2013.01.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/27/2012] [Accepted: 12/07/2012] [Indexed: 10/27/2022]
Abstract
Placental malaria (PM) is a major cause of fetal growth restriction, yet the underlying mechanism is unclear. Complement C5a and C5a receptor levels are increased with PM. C5a is implicated in fetal growth restriction in non-infection-based animal models. In a case-control study of 492 pregnant Malawian women, we find that elevated C5a levels are associated with an increased risk of delivering a small-for-gestational-age infant. C5a was significantly increased in PM and was negatively correlated with the angiogenic factor angiopoietin-1 and positively correlated with angiopoietin-2, soluble endoglin, and vascular endothelial growth factor. Genetic or pharmacological blockade of C5a or its receptor in a mouse model of PM resulted in greater fetoplacental vessel development, reduced placental vascular resistance, and improved fetal growth and survival. These data suggest that C5a drives fetal growth restriction in PM through dysregulation of angiogenic factors essential for placental vascular remodeling resulting in placental vascular insufficiency.
Collapse
Affiliation(s)
- Andrea L Conroy
- Sandra Rotman Laboratories, Sandra Rotman Centre, University Health Network-Toronto General Hospital, University of Toronto, Toronto, ON M5G 1L7, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|