1
|
Garcia-Flores V, Liu Z, Romero R, Pique-Regi R, Xu Y, Miller D, Levenson D, Galaz J, Winters AD, Farias-Jofre M, Panzer JJ, Theis KR, Gomez-Lopez N. Homeostatic Macrophages Prevent Preterm Birth and Improve Neonatal Outcomes by Mitigating In Utero Sterile Inflammation in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1620-1634. [PMID: 39431882 PMCID: PMC11572957 DOI: 10.4049/jimmunol.2400467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/28/2024] [Indexed: 10/22/2024]
Abstract
Preterm birth (PTB), often preceded by preterm labor, is a major cause of neonatal morbidity and mortality worldwide. Most PTB cases involve intra-amniotic inflammation without detectable microorganisms, termed in utero sterile inflammation, for which there is no established treatment. In this study, we propose homeostatic macrophages to prevent PTB and adverse neonatal outcomes caused by in utero sterile inflammation. Single-cell atlases of the maternal-fetal interface revealed that homeostatic maternal macrophages are reduced with human labor. M2 macrophage treatment prevented PTB and reduced adverse neonatal outcomes in mice with in utero sterile inflammation. Specifically, M2 macrophages halted premature labor by suppressing inflammatory responses in the amniotic cavity, including inflammasome activation, and mitigated placental and offspring lung inflammation. Moreover, M2 macrophages boosted gut inflammation in neonates and improved their ability to fight systemic bacterial infections. Our findings show that M2 macrophages are a promising strategy to mitigate PTB and improve neonatal outcomes resulting from in utero sterile inflammation.
Collapse
Affiliation(s)
- Valeria Garcia-Flores
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Zhenjie Liu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
| | - Roberto Romero
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI
| | - Roger Pique-Regi
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI
| | - Yi Xu
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Derek Miller
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
| | - Dustyn Levenson
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI
| | - Jose Galaz
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrew D. Winters
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Marcelo Farias-Jofre
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Division of Obstetrics and Gynecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jonathan J. Panzer
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Kevin R. Theis
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
| | - Nardhy Gomez-Lopez
- Pregnancy Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI and Bethesda, MD
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI
- Center for Reproductive Health Sciences, Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
2
|
Carvajal JA, Galaz J, Villagrán S, Astudillo R, Garmendia L, Delpiano AM. The role of the RHOA/ROCK pathway in the regulation of myometrial stages throughout pregnancy. AJOG GLOBAL REPORTS 2024; 4:100394. [PMID: 39434813 PMCID: PMC11491706 DOI: 10.1016/j.xagr.2024.100394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024] Open
Abstract
Background Controlling uterine contractile activity is essential to regulate the duration of pregnancy. During most of the pregnancy, the uterus does not contract (i.e., myometrial quiescence). The myometrium recovers its contractile phenotype at around 36 weeks (i.e., myometrial activation) through several mechanisms. The RHOA/ROCK pathway plays a vital role in facilitating muscular contractions by calcium sensitization in humans. Yet, the role of this pathway during different myometrial stages, including quiescence, has not been elucidated. Objective we aimed to study the role of the RHOA/ROCK pathway in the regulation of the different myometrial stages throughout pregnancy. Specifically, we hypothesized that the inhibition of the components of the RHOA/ROCK pathway play an important role in maintaining uterine quiescence. Study design Myometrial samples were obtained from pregnant individuals who underwent cesarean section. Pregnant individuals who delivered preterm without labor (myometrial quiescence), preterm with labor (nonphysiological myometrial stimulation), term not in labor (activation), and term in labor (physiological myometrial stimulation) were included. The mRNA and protein expression of RHOA, ROCK I, ROCK II, RND1-3, and ROCK activity through pMYTP1 were evaluated. Results We found that the human myometrium constitutively expressed RHOA/ROCK pathway components throughout pregnancy. No changes in the components of the RHOA/ROCK pathway were found during quiescence. Moreover, the RHOA protein and ROCK activity increased in the myometrium during labor, supporting the hypothesis that this pathway participates in maintaining the contractile activity of the myometrium. This study provides insight into the role of the RHOA/ROCK pathway in controlling myometrial contractile activity during pregnancy.
Collapse
Affiliation(s)
- Jorge A. Carvajal
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| | - Jose Galaz
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| | - Sofía Villagrán
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| | - Rocío Astudillo
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| | - Liliana Garmendia
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| | - Ana María Delpiano
- Departamento de Obstetricia, Escuela de Medicina, Unidad de Medicina Materno Fetal, Pontificia Universidad Católica de Chile, Santiago, Chile (All authors)
| |
Collapse
|
3
|
Zambuto SG, Kolluru SS, Harley BAC, Oyen ML. Gelatin methacryloyl biomaterials and strategies for trophoblast research. Placenta 2024; 157:67-75. [PMID: 39341721 DOI: 10.1016/j.placenta.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Rising maternal mortality rates in the U.S. are a significant public health issue that must be addressed; however, much of the basic science information required to target pregnancy-related pathologies have not yet been defined. Placental and blastocyst implantation research are challenging to perform in humans because of the early time frame of these processes in pregnancy and limited access to first trimester tissues. As a result, there is a critical need to develop model systems capable of studying these processes in increasing mechanistic detail. With the recent passing of the FDA Modernization Act 2.0 and advances in tissue engineering methods, three-dimensional microphysiological model systems offer an exciting opportunity to model early stages of placentation. Here, we detail the synthesis, characterization, and application of gelatin methacryloyl (GelMA) hydrogel platforms for studying trophoblast behavior in three-dimensional hydrogel systems. Photopolymerization strategies to fabricate GelMA hydrogels render the hydrogels homogeneous in terms of structure and stable under physiological temperatures, allowing for rigorous fabrication of reproducible hydrogel variants. Unlike other natural polymers that have minimal opportunity to tune their properties, GelMA hydrogel properties can be tuned across many axes of variation, including polymer degree of functionalization, gelatin bloom strength, light exposure time and intensity, polymer weight percent, photoinitiator concentration, and physical geometry. In this work, we aim to inspire and instruct the field to utilize GelMA biomaterial strategies for future placental research. With enhanced microphysiological models of pregnancy, we can now generate the basic science information required to address problems in pregnancy.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63130, USA; Dept. of Biomedical Engineering, USA; Center for Women's Health Engineering, USA.
| | - Samyuktha S Kolluru
- Center for Women's Health Engineering, USA; The Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Michelle L Oyen
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63130, USA; Dept. of Biomedical Engineering, USA; Center for Women's Health Engineering, USA; The Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| |
Collapse
|
4
|
Curtis MW, Lopez JE. Tick-Borne Diseases and Pregnancy: A Narrative Review Evaluating Pregnancy Complications Caused by Tick-Borne Diseases. Trop Med Infect Dis 2024; 9:254. [PMID: 39591260 PMCID: PMC11598240 DOI: 10.3390/tropicalmed9110254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 11/28/2024] Open
Abstract
Ticks are vectors of public health concern because the pathogens they transmit can cause detrimental diseases in humans. Lyme disease, tick-borne relapsing fever, human granulocytic anaplasmosis, Rocky Mountain spotted fever, tick-borne encephalitis, Crimean-Congo hemorrhagic fever, and babesiosis are some of the most common diseases caused by the pathogens transmitted by ticks. The overlap between human activities and tick habitats is growing, contributing to an increase in tick-borne disease cases. Unfortunately, pregnancy as a risk factor for tick-borne diseases is largely ignored. In this narrative review we use case reports, epidemiological studies, and animal studies to evaluate the maternal, pregnancy, and fetal outcomes caused by Lyme disease, tick-borne relapsing fever, human granulocytic anaplasmosis, Rocky Mountain spotted fever, tick-borne encephalitis, Crimean-Congo hemorrhagic fever, and babesiosis during pregnancy.
Collapse
Affiliation(s)
- Michael W. Curtis
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Job E. Lopez
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Wray S, Taggart MJ. An update on pacemaking in the myometrium. J Physiol 2024. [PMID: 39073139 DOI: 10.1113/jp284753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/24/2024] [Indexed: 07/30/2024] Open
Abstract
Timely and efficient contractions of the smooth muscle of the uterus - the myometrium - are crucial to a successful pregnancy outcome. These episodic contractions are regulated by spontaneous action potentials changing cell and tissue electrical excitability. In this short review we will document and discuss current knowledge of these processes. Those seeking a conclusive account of myometrial pacemaking mechanisms, or indeed a definitive description of the anatomical site of uterine pacemaking, may be disappointed. Rather, after almost a century of investigation, and in spite of promising studies in the last decade or so, there remain many gaps in our knowledge. We review the progress that has been made using recent technologies including in vivo and ex vivo imaging and electrophysiology and computational modelling, taking evidence from studies of animal and human myometrium, with particular emphasis on what may occur in the latter. We have prioritized physiological studies that bring us closer to understanding function. From our analyses we suggest that in human myometrium there is no fixed pacemaking site, but rather mobile, initiation sites produce the connectivity for synchronizing electrical and contractile activity. We call for more studies and funding, as physiological understanding of pacemaking gives hope to being better able to treat clinical conditions such as preterm and dysfunctional labours.
Collapse
Affiliation(s)
- Susan Wray
- Women's & Children's Health, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, Merseyside, UK
| | - Michael J Taggart
- Biosciences Institute, International Centre for Life, Newcastle University, Newcastle, UK
| |
Collapse
|
6
|
Cortes DRE, Stapleton MC, Schwab KE, West D, Coulson NW, O’Donnell MG, Christodoulou AG, Powers RW, Wu YL. Modeling normal mouse uterine contraction and placental perfusion with non-invasive longitudinal dynamic contrast enhancement MRI. PLoS One 2024; 19:e0303957. [PMID: 38950083 PMCID: PMC11216620 DOI: 10.1371/journal.pone.0303957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 05/05/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND The placenta is a transient organ critical for fetal development. Disruptions of normal placental functions can impact health throughout an individual's entire life. Although being recognized by the NIH Human Placenta Project as an important organ, the placenta remains understudied, partly because of a lack of non-invasive tools for longitudinally evaluation for key aspects of placental functionalities. OBJECTIVE Our goal is to create a non-invasive preclinical imaging pipeline that can longitudinally probe murine placental health in vivo. We use advanced imaging processing schemes to establish functional biomarkers for non-invasive longitudinal evaluation of placental development. METHODOLOGY We implement dynamic contrast enhancement magnetic resonance imaging (DCE-MRI) and analysis pipeline to quantify uterine contraction and placental perfusion dynamics. We use optic flow and time-frequency analysis to quantify and characterize contraction-related placental motion. Our novel imaging and analysis pipeline uses subcutaneous administration of gadolinium for steepest slope-based perfusion evaluation, enabling non-invasive longitudinal monitoring. RESULTS We demonstrate that the placenta exhibits spatially asymmetric contractile motion that develops from E14.5 to E17.5. Additionally, we see that placental perfusion, perfusion delivery rate, and substrate delivery all increase from E14.5 to E17.5, with the High Perfusion Chamber (HPC) leading the placental changes that occur from E14.5 to E17.5. DISCUSSION We advance the placental perfusion chamber paradigm with a novel, physiologically based threshold model for chamber localization and demonstrate spatially varying placental chambers using multiple functional metrics that assess mouse placental development and remodeling throughout gestation. CONCLUSION Our pipeline enables the non-invasive, longitudinal assessment of multiple placenta functions from a single imaging session. Our pipeline serves as a key toolbox for advancing research in mouse models of placental disease and disorder.
Collapse
Affiliation(s)
- Devin Raine Everaldo Cortes
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Margaret C. Stapleton
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kristina E. Schwab
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| | - Dalton West
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Noah W. Coulson
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | - Anthony G. Christodoulou
- Department of Radiological Sciences and Engineering, University of California, Los Angeles, California, United States of America
| | - Robert W. Powers
- Magee-Womens Research Institute, Pittsburgh, PA, United States of America
| | - Yijen L. Wu
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA, United States of America
- Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
7
|
Kaissar MS, Yoshida K. Computational model captures cardiac growth in hypertensive pregnancies and in the postpartum period. Am J Physiol Heart Circ Physiol 2024; 326:H1491-H1497. [PMID: 38668702 PMCID: PMC11380950 DOI: 10.1152/ajpheart.00104.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/30/2024]
Abstract
Heart growth in the pregnant patient helps maintain cardiovascular function while supporting the growing fetus. However, in some cases, the cardiovascular demand of pregnancy can trigger life-threatening conditions, including hypertensive disorders of pregnancy and peripartum cardiomyopathy. The mechanisms that control heart growth throughout pregnancy are unclear, and treating these diseases remains elusive. We previously developed a computational model that accounts for hormonal and hemodynamic interactions throughout pregnancy and demonstrated its ability to capture realistic cardiac growth in normal rat pregnancy. In this study, we evaluated whether this model could capture heart growth beyond normal pregnancy. After further validation of our normal pregnancy predictions, we tested our model predictions of three rat studies of hypertensive pregnancies. We next simulated the postpartum period and examined the impact of lactation on cardiac growth in rats. We demonstrate that our multiscale model can capture cardiac growth associated with new-onset hypertension during pregnancy and lactation status in the postpartum period. We conclude by elaborating on the potential clinical utility of our model in the future.NEW & NOTEWORTHY Our multiscale model predicts appropriate heart growth beyond normal pregnancy, including elevated heart weights in rats with induced hypertension during pregnancy and in lactating mice and decreased heart weight in nonlactating mice. Our model captures distinct mechanisms that result in similar organ-level growth, highlighting its potential to distinguish healthy from diseased pregnancy-induced growth.
Collapse
Affiliation(s)
- Molly S Kaissar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States
| | - Kyoko Yoshida
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
8
|
Cortes DRE, Stapleton MC, Schwab KE, West D, Coulson NW, O'Donnell MG, Powers RW, Wu YL. Modeling Normal Mouse Uterine Contraction and Placental Perfusion with Non-invasive Longitudinal Dynamic Contrast Enhancement MRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.577398. [PMID: 38352563 PMCID: PMC10862875 DOI: 10.1101/2024.01.31.577398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2024]
Abstract
The placenta is a transient organ critical for fetal development. Disruptions of normal placental functions can impact health throughout an individual's entire life. Although being recognized by the NIH Human Placenta Project as an important organ, the placenta remains understudied, partly because of a lack of non-invasive tools for longitudinally evaluation for key aspects of placental functionalities. Non-invasive imaging that can longitudinally probe murine placental health in vivo are critical to understanding placental development throughout pregnancy. We developed advanced imaging processing schemes to establish functional biomarkers for non-invasive longitudinal evaluation of placental development. We developed a dynamic contrast enhancement magnetic resonance imaging (DCE-MRI) pipeline combined with advanced image process methods to model uterine contraction and placental perfusion dynamics. Our novel imaging pipeline uses subcutaneous administration of gadolinium for steepest-slope based perfusion evaluation. This enables non-invasive longitudinal monitoring. Additionally, we advance the placental perfusion chamber paradigm with a novel physiologically-based threshold model for chamber localization and demonstrate spatially varying placental chambers using multiple functional metrics that assess mouse placental development and continuing remodeling throughout gestation. Lastly, using optic flow to quantify placental motions arisen from uterine contractions in conjunction with time-frequency analysis, we demonstrated that the placenta exhibited asymmetric contractile motion.
Collapse
Affiliation(s)
- Devin Raine Everaldo Cortes
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh, PA
| | - Margaret C Stapleton
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh, PA
| | - Kristina E Schwab
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA
| | - Dalton West
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
| | - Noah W Coulson
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Yijen L Wu
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA
- Rangos Research Center Animal Imaging Core, Children's Hospital of Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
9
|
Omotehara T, Nakata H, Nagahori K, Kuramasu M, Ichimura K, Itoh M. A Single Administration of Progesterone during the Neonatal Period Shows No Structural Changes in Male Reproductive Tracts in Mice. Acta Histochem Cytochem 2023; 56:127-136. [PMID: 38318101 PMCID: PMC10838630 DOI: 10.1267/ahc.23-00052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/10/2023] [Indexed: 02/07/2024] Open
Abstract
The concentration of female-dominant steroid hormones, such as progesterone and estrogen, drops after birth in neonates. We have reported that neonatal estrogen treatment results in inflammation in the epididymis after puberty in male mice. Our recent study discovered that progesterone receptor was specifically expressed in efferent ducts just before birth in male mice. Therefore, this study aimed to reveal the impact of neonatal progesterone administration on the efferent ducts after puberty. Progesterone was subcutaneously administered to neonatal mice on their birthday in three groups: high-dose (200 mg/kg), low-dose (8 mg/kg), and control (cottonseed oil). Their testis and epididymis were collected at 12 weeks old. Semi-serial paraffin sections of these tissues were prepared and evaluated through PAS-hematoxylin staining. Efferent ducts were reconstructed into a three-dimensional structure, and their length and volume were analyzed. Spermatogenesis in the testis and epithelium of the tracts appeared normal, even in individuals administered with progesterone. There were no significant differences in the length and volume of the efferent ducts among the three groups. This study suggests that progesterone treatment in neonatal mice does not cause any structural changes in the male reproductive tracts at puberty, unlike the neonatal estrogen treatment.
Collapse
Affiliation(s)
- Takuya Omotehara
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Hiroki Nakata
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Ishikawa, Japan
| | - Kenta Nagahori
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
- Division of Basic Medical Science, Department of Anatomy, Tokai University School of Medicine, Kanagawa, Japan
| | - Miyuki Kuramasu
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Koichiro Ichimura
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masahiro Itoh
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
10
|
Shynlova O, Nadeem L, Lye S. Progesterone control of myometrial contractility. J Steroid Biochem Mol Biol 2023; 234:106397. [PMID: 37683774 DOI: 10.1016/j.jsbmb.2023.106397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/01/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
During pregnancy, the primary function of the uterus is to be quiescent and not contract, which allows the growing fetus to develop and mature. A uterine muscle layer, myometrium, is composed of smooth muscle cells (SMCs). Before the onset of labor contractions, the uterine SMCs experience a complex biochemical and molecular transformation involving the expression of contraction-associated proteins. Labor is initiated when genes in SMCs are activated in response to a combination of hormonal, inflammatory and mechanical signals. In this review, we provide an overview of molecular mechanisms regulating the process of parturition in humans, focusing on the hormonal control of the myometrium, particularly the steroid hormone progesterone. The primary reason for discussing the regulation of myometrial contractility by progesterone is the importance of the clinical problem of preterm birth. It is thought that the hormonal mechanisms regulating premature uterine contractions represent an untimely triggering of the normal events occurring during term parturition. Yet, our knowledge of the complex and redundant hormonal pathways controlling uterine contractile activity leading to delivery of the neonate remains incomplete. Finally, we introduce recent animal studies using a novel class of drugs, Selective Progesterone Receptor Modulators, targeting progesterone signaling to prevent premature myometrial contractions.
Collapse
Affiliation(s)
- Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada.
| | - Lubna Nadeem
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada
| | - Stephen Lye
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto M5G 1X5, Canada; Department of Physiology, University of Toronto, M5S 1A1, Canada; Department of Obstetrics & Gynecology, University of Toronto, M5S 1A1, Canada
| |
Collapse
|
11
|
Maxey AP, Travis JM, McCain ML. Regulation of oxytocin-induced calcium transients and gene expression in engineered myometrial tissues by tissue architecture and matrix rigidity. Curr Res Physiol 2023; 6:100108. [PMID: 38107790 PMCID: PMC10724203 DOI: 10.1016/j.crphys.2023.100108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/21/2023] [Accepted: 09/20/2023] [Indexed: 12/19/2023] Open
Abstract
The uterus is susceptible to benign tumors known as fibroids, which have been associated with many pregnancy complications, including preterm labor. However, the impact of fibrotic tissue remodeling on the physiology of the myometrium, the smooth muscle layer of the uterus, is poorly understood, in large part due to a lack of model systems. In this study, we engineered healthy-like and fibrotic-like myometrium by culturing human myometrial smooth muscle cells on polyacrylamide hydrogels micropatterned with fibronectin to independently tune matrix rigidity and tissue alignment, respectively. We then evaluated calcium transients in response to oxytocin stimulation. Isotropic myometrial tissues on stiff substrates (representing fibrotic myometrium) had shorter calcium transients due to shorter decay time compared to aligned myometrial tissues on soft substrates (representing healthy myometrium). Calcium transients in aligned tissues had longer response times and longer decay times than isotropic tissues, irrespective of substrate stiffness. The amplitude of calcium transients was also higher on soft substrates compared to stiff substrates, irrespective of tissue alignment. We also performed RNA sequencing to detect differentially expressed genes between healthy- and fibrotic-like tissues, which revealed that a bitter taste receptor shown to induce smooth muscle relaxation, TAS2R31, was down-regulated in fibrotic-like tissues. Finally, we measured oxytocin-induced calcium transients in response to pre-treatment with progesterone, caffeine, thrombin, and nifedipine to demonstrate applications for our model system in drug screening. Both progesterone and caffeine caused a decrease in calcium transient duration, as expected, while thrombin and nifedipine had less impact. Collectively, our engineered model of the myometrium enables new insights into myometrial mechanobiology and can be extended to identify or screen novel drug targets.
Collapse
Affiliation(s)
- Antonina P. Maxey
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Jaya M. Travis
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Megan L. McCain
- Laboratory for Living Systems Engineering, Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
12
|
Schuler G. [Initiation and endocrine control of parturition in domestic mammals - Part 1]. Tierarztl Prax Ausg G Grosstiere Nutztiere 2023; 51:228-236. [PMID: 37820628 DOI: 10.1055/a-2142-3318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Endocrine regulation of parturition is based on an intense exchange of signals between the fetus, placenta and mother. Apart from sheep, our knowledge of the endocrine control of parturition is still very incomplete. However, current observations suggest significant differences between the species. For the maintenance of pregnancy, progesterone (P4) is the crucial superordinate regulatory factor, although in some species, such as the horse, functions of P4 are at least partially fulfilled by other progestogens. In general, prepartum P4 withdrawal is considered a prerequisite for the onset of physiological birth. In species with exclusive (dog) or predominant (e. g., cattle, goat, pig) luteal P4 at the end of gestation, luteolysis is the crucial event. In sheep, where P4 is of placental origin prior to parturition, the prepartum P4 decline is due to a switch in placental steroid metabolism. The mechanism of prepartum progestogen withdrawal in the mare is still largely unclear. In sheep, initiation of parturition proceeds from maturation of the fetal hypothalamic-pituitary-adrenal (HPA) axis, which leads to a steep prepartum rise in fetal cortisol concentrations stimulating the collapse of placental P4 production. In cattle, fetal cortisol probably triggers luteolysis via stimulation of placental prostaglandin secretion. In several other domestic mammalian species, there is also evidence that the initiation of parturition proceeds from maturation of the fetal HPA axis. However, the functional relationships between fetal cortisol and prepartum P4 withdrawal are largely unknown in nonruminant species.
Collapse
Affiliation(s)
- Gerhard Schuler
- Tierklinik für Reproduktionsmedizin und Neugeborenenkunde, Justus-Liebig-Universität Giessen
| |
Collapse
|
13
|
Joseph A, Lewis EL, Ferguson B, Guan Y, Anton L, Elovitz MA. Intrauterine colonization with Gardnerella vaginalis and Mobiluncus mulieris induces maternal inflammation but not preterm birth in a mouse model. Am J Reprod Immunol 2023; 90:e13749. [PMID: 37491927 PMCID: PMC11423284 DOI: 10.1111/aji.13749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/27/2023] Open
Abstract
PROBLEM Preterm birth (PTB) remains a leading cause of childhood mortality. Recent studies demonstrate that the risk of spontaneous PTB (sPTB) is increased in individuals with Lactobacillus-deficient vaginal microbial communities. One proposed mechanism is that vaginal microbes ascend through the cervix, colonize the uterus, and activate inflammatory pathways leading to sPTB. This study assessed whether intrauterine colonization with either Gardnerella vaginalis and Mobiluncus mulieris alone is sufficient to induce maternal-fetal inflammation and induce sPTB. METHOD OF STUDY C56/B6J mice, on embryonic day 15, received intrauterine inoculation of saline or 108 colony-forming units of G. vaginalis (n = 30), M. mulieris (n = 17), or Lactobacillus crispatus (n = 16). Dams were either monitored for maternal morbidity and sPTB or sacrificed 6 h post-infusion for analysis of bacterial growth and cytokine/chemokine expression in maternal and fetal tissues. RESULTS Six hours following intrauterine inoculation with G. vaginalis, M. mulieris, or L. crispatus, live bacteria were observed in both blood and amniotic fluid, and a potent immune response was identified in the uterus and maternal serum. In contrast, only a limited immune response was identified in the amniotic fluid and the fetus after intrauterine inoculation. High bacterial load (108 CFU/animal) of G. vaginalis was associated with maternal morbidity and mortality but not sPTB. Intrauterine infusion with L. crispatus or M. mulieris at 108 CFU/animal did not induce sPTB, alter pup viability, litter size, or maternal mortality. CONCLUSIONS Despite inducing an immune response, intrauterine infusion of live G. vaginalis or M. mulieris is not sufficient to induce sPTB in our mouse model. These results suggest that ascension of common vaginal microbes into the uterine cavity alone is not causative for sPTB.
Collapse
Affiliation(s)
- Andrea Joseph
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine, New York, New York, USA
| | - Emma L Lewis
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Briana Ferguson
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yuxia Guan
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Lauren Anton
- Center for Research on Reproduction and Women's Health, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michal A Elovitz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Icahn School of Medicine, New York, New York, USA
| |
Collapse
|
14
|
Sun J, Chong J, Zhang J, Ge L. Preterm pigs for preterm birth research: reasonably feasible. Front Physiol 2023; 14:1189422. [PMID: 37520824 PMCID: PMC10374951 DOI: 10.3389/fphys.2023.1189422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 07/07/2023] [Indexed: 08/01/2023] Open
Abstract
Preterm birth will disrupt the pattern and course of organ development, which may result in morbidity and mortality of newborn infants. Large animal models are crucial resources for developing novel, credible, and effective treatments for preterm infants. This review summarizes the classification, definition, and prevalence of preterm birth, and analyzes the relationship between the predicted animal days and one human year in the most widely used animal models (mice, rats, rabbits, sheep, and pigs) for preterm birth studies. After that, the physiological characteristics of preterm pig models at different gestational ages are described in more detail, including birth weight, body temperature, brain development, cardiovascular system development, respiratory, digestive, and immune system development, kidney development, and blood constituents. Studies on postnatal development and adaptation of preterm pig models of different gestational ages will help to determine the physiological basis for survival and development of very preterm, middle preterm, and late preterm newborns, and will also aid in the study and accurate optimization of feeding conditions, diet- or drug-related interventions for preterm neonates. Finally, this review summarizes several accepted pediatric applications of preterm pig models in nutritional fortification, necrotizing enterocolitis, neonatal encephalopathy and hypothermia intervention, mechanical ventilation, and oxygen therapy for preterm infants.
Collapse
Affiliation(s)
- Jing Sun
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Jie Chong
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
| | - Jinwei Zhang
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| | - Liangpeng Ge
- Chongqing Academy of Animal Sciences, Chongqing, China
- National Center of Technology Innovation for Pigs, Chongqing, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Chongqing, China
| |
Collapse
|
15
|
Valerio E, Stocchero M, Pirillo P, D'Errico I, Bonadies L, Galderisi A, Giordano G, Baraldi E. Neurosteroid pathway derangement in asphyctic infants treated with hypothermia: an untargeted metabolomic approach. EBioMedicine 2023; 92:104636. [PMID: 37257315 PMCID: PMC10244906 DOI: 10.1016/j.ebiom.2023.104636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/29/2023] [Accepted: 05/15/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND The pathobiological mechanisms associated with perinatal asphyxia and hypoxic-ischemic encephalopathy are complex and poorly understood. The metabolic effects of therapeutic hypothermia have been partially explored. METHODS We conducted a single-center longitudinal study to investigate the metabolic effects of perinatal asphyxia and hypoxic-ischemic encephalopathy on the urinary metabolome of a group of 12 asphyctic infants over time compared to 22 matched healthy newborns, using untargeted metabolomics based on mass spectrometry. FINDINGS Over-representation pathway analysis identified the steroidogenesis pathway as being significantly disrupted, with reduced steroid levels in the first three days of life despite treatment with hypothermia. Comparison with matched healthy newborns showed that the urinary steroid content was lower in asphyctic infants before hypothermia. The lysine degradation and carnitine synthesis pathways were also significantly affected. INTERPRETATION Steroidogenesis is significantly disrupted in asphyctic infants compared to healthy newborns. Given how neurosteroids are involved in neuromodulation and neuroprotection, translational research is warranted on the potential role of neurosteroid-based intervention in asphyctic infants. FUNDING None.
Collapse
Affiliation(s)
- Enrico Valerio
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy
| | - Matteo Stocchero
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy
| | - Paola Pirillo
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy
| | - Ignazio D'Errico
- Department of Neuroradiology, Azienda Ospedale-Università di Padova, Italy
| | - Luca Bonadies
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy
| | - Alfonso Galderisi
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Department of Pediatrics, Yale School of Medicine, Yale University, New Haven, CT 06511, USA
| | - Giuseppe Giordano
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Woman's and Child's Health, Azienda Ospedale-Università di Padova, Italy; Institute of Pediatric Research "Città Della Speranza", Padova, Italy.
| |
Collapse
|
16
|
Stock SJ, Aiken CE. Barriers to progress in pregnancy research: How can we break through? Science 2023; 380:150-153. [PMID: 37053324 DOI: 10.1126/science.adf9347] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/24/2023] [Indexed: 04/15/2023]
Abstract
Healthy pregnancies are fundamental to healthy populations, but very few therapies to improve pregnancy outcomes are available. Fundamental concepts-for example, placentation or the mechanisms that control the onset of labor-remain understudied and incompletely understood. A key issue is that research efforts must capture the complexity of the tripartite maternal-placental-fetal system, the dynamics of which change throughout gestation. Studying pregnancy disorders is complicated by the difficulty of creating maternal-placental-fetal interfaces in vitro and the uncertain relevance of animal models to human pregnancy. However, newer approaches include trophoblast organoids to model the developing placenta and integrated data-science approaches to study longer-term outcomes. These approaches provide insights into the physiology of healthy pregnancy, which is the first step to identifying therapeutic targets in pregnancy disorders.
Collapse
Affiliation(s)
- Sarah J Stock
- University of Edinburgh Usher Institute, Edinburgh EH16 4UX, UK
- University of Edinburgh MRC Centre for Reproductive Health, Edinburgh EH16 4TJ, UK
- Wellcome Leap In Utero Program, Wellcome Leap Inc., Culver City, CA 90232, USA
| | - Catherine E Aiken
- The Rosie Hospital and NIHR Cambridge Biomedical Research Centre, Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0SW, UK
- Centre for Trophoblast Research, Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0SW, UK
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
17
|
Candia AA, Jiménez T, Navarrete Á, Beñaldo F, Silva P, García-Herrera C, Sferruzzi-Perri AN, Krause BJ, González-Candia A, Herrera EA. Developmental Ultrasound Characteristics in Guinea Pigs: Similarities with Human Pregnancy. Vet Sci 2023; 10:144. [PMID: 36851448 PMCID: PMC9963037 DOI: 10.3390/vetsci10020144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Biometrical and blood flow examinations are fundamental for assessing fetoplacental development during pregnancy. Guinea pigs have been proposed as a good model to study fetal development and related gestational complications; however, longitudinal growth and blood flow changes in utero have not been properly described. This study aimed to describe fetal and placental growth and blood flow of the main intrauterine vascular beds across normal guinea pig pregnancy and to discuss the relevance of this data for human pregnancy. METHODS Pregnant guinea pigs were studied from day 25 of pregnancy until term (day ~70) by ultrasound and Doppler assessment. The results were compared to human data from the literature. RESULTS Measurements of biparietal diameter (BPD), cranial circumference (CC), abdominal circumference, and placental biometry, as well as pulsatility index determination of umbilical artery, middle cerebral artery (MCA), and cerebroplacental ratio (CPR), were feasible to determine across pregnancy, and they could be adjusted to linear or nonlinear functions. In addition, several of these parameters showed a high correlation coefficient and could be used to assess gestational age in guinea pigs. We further compared these data to ultrasound variables from human pregnancy with high similarities. CONCLUSIONS BPD and CC are the most reliable measurements to assess fetal growth in guinea pigs. Furthermore, this is the first report in which the MCA pulsatility index and CPR are described across guinea pig gestation. The guinea pig is a valuable model to assess fetal growth and blood flow distribution, variables that are comparable with human pregnancy.
Collapse
Affiliation(s)
- Alejandro A. Candia
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Tamara Jiménez
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Álvaro Navarrete
- Departamento de Ingeniería Mecánica, Facultad de Ingeniería, Universidad de Santiago de Chile, Santiago 9170022, Chile
| | - Felipe Beñaldo
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Pablo Silva
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
| | - Claudio García-Herrera
- Departamento de Ingeniería Mecánica, Facultad de Ingeniería, Universidad de Santiago de Chile, Santiago 9170022, Chile
| | - Amanda N. Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3EG, UK
| | - Bernardo J. Krause
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Alejandro González-Candia
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Institute of Health Sciences, University of O’Higgins, Rancagua 2841959, Chile
| | - Emilio A. Herrera
- Laboratorio de Función y Reactividad Vascular, Programa de Fisiopatología, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- International Center for Andean Studies (INCAS), University of Chile, Putre 1070000, Chile
| |
Collapse
|
18
|
Miller FA, Sacco A, David AL, Boyle AK. Interventions for Infection and Inflammation-Induced Preterm Birth: a Preclinical Systematic Review. Reprod Sci 2023; 30:361-379. [PMID: 35426035 PMCID: PMC9988807 DOI: 10.1007/s43032-022-00934-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/02/2022] [Indexed: 12/09/2022]
Abstract
Spontaneous preterm births (< 37 weeks gestation) are frequently associated with infection. Current treatment options are limited but new therapeutic interventions are being developed in animal models. In this PROSPERO-registered preclinical systematic review, we aimed to summarise promising interventions for infection/inflammation-induced preterm birth. Following PRISMA guidance, we searched PubMed, EMBASE, and Web of Science using the themes: "animal models", "preterm birth", "inflammation", and "therapeutics". We included original quantitative, peer-reviewed, and controlled studies applying prenatal interventions to prevent infection/inflammation-induced preterm birth in animal models. We employed two risk of bias tools. Of 4020 identified studies, 23 studies (24 interventions) met our inclusion criteria. All studies used mouse models. Preterm birth was most commonly induced by lipopolysaccharide (18 studies) or Escherichia coli (4 studies). Models varied according to infectious agent serotype, dose, and route of delivery. Gestational length was significantly prolonged in 20/24 interventions (83%) and markers of maternal inflammation were reduced in 20/23 interventions (87%). Interventions targeting interleukin-1, interleukin-6, and toll-like receptors show particular therapeutic potential. However, due to the heterogeneity of the methodology of the included studies, meta-analysis was impossible. All studies were assigned an unclear risk of bias using the SYRCLE risk of bias tool. Interventions targeting inflammation demonstrate therapeutic potential for the prevention of preterm birth. However, better standardisation of preterm birth models, including the dose, serotype, timing of administration and pathogenicity of infectious agent, and outcome reporting is urgently required to improve the reproducibility of preclinical studies, allow meaningful comparison of intervention efficacy, and aid clinical translation.
Collapse
Affiliation(s)
- Faith A Miller
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Adalina Sacco
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
| | - Anna L David
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK
- National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, UK
| | - Ashley K Boyle
- Elizabeth Garrett Anderson Institute for Women's Health, University College London, 86-96 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
19
|
Kyathanahalli C, Snedden M, Hirsch E. Is human labor at term an inflammatory condition?†. Biol Reprod 2023; 108:23-40. [PMID: 36173900 PMCID: PMC10060716 DOI: 10.1093/biolre/ioac182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 01/20/2023] Open
Abstract
Parturition at term in normal pregnancy follows a predictable sequence of events. There is some evidence that a state of inflammation prevails in the reproductive tissues during labor at term, but it is uncertain whether this phenomenon is the initiating signal for parturition. The absence of a clear temporal sequence of inflammatory events prior to labor casts doubt on the concept that normal human labor at term is primarily the result of an inflammatory cascade. This review examines evidence linking parturition and inflammation in order to address whether inflammation is a cause of labor, a consequence of labor, or a separate but related phenomenon. Finally, we identify and suggest ways to reconcile inconsistencies regarding definitions of labor onset in published research, which may contribute to the variability in conclusions regarding the genesis and maintenance of parturition. A more thorough understanding of the processes underlying normal parturition at term may lead to novel insights regarding abnormal labor, including spontaneous preterm labor, preterm premature rupture of the fetal membranes, and dysfunctional labor, and the role of inflammation in each.
Collapse
Affiliation(s)
- Chandrashekara Kyathanahalli
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Madeline Snedden
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
| | - Emmet Hirsch
- Department of Obstetrics and Gynecology, NorthShore University HealthSystem, Evanston, Illinois, USA
- Department of Obstetrics and Gynecology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
Birchenall KA, Welsh GI, López Bernal A. The feto-placental metabolome of spontaneous labour is not reproduced following induction of labour. Placenta 2022; 129:111-118. [PMID: 36288646 DOI: 10.1016/j.placenta.2022.10.005] [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: 01/10/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The mechanism for human labour remains poorly understood, limiting our ability to manage complications including spontaneous preterm birth and induction of labour (IOL). The study of fetal signals poses specific challenges. Metabolomic analysis of maternal blood, the cord artery (CA), and cord vein (CV), allows simultaneous interrogation of multiple metabolic pathways associated with different modes of labour onset and birth. METHODS Global mass spectrometry metabolomics analysis was performed on serial samples collected from participants during pregnancy, in latent phase of labour, and following birth (CA, CV, and intervillous (IV) blood), from those who spontaneously laboured and birthed vaginally (SL group), had IOL and birthed vaginally (IOL group), or birthed via elective caesarean section (no labour; ECS group). RESULTS There were clear differences in fetal and maternal steroid, arachidonate and sphingosine pathways between the SL and IOL groups, despite similar uterine contractions and vaginal birth. The CA/CV ratio for key steroids of the IOL group were more alike the ECS group than the SL group, including progesterone (CA/CV ratio for: SL group=3.5; IOL group=0.5; and ECS group=0.5), and oestriol (CA/CV ratio for: SL group=4.3; IOL group=0.4; and for ECS group=0.2). There were no such changes in the maternal samples. DISCUSSION These findings indicate that IOL does not reproduce the pathways activated in spontaneous labour. The decreased placental progesterone production observed with spontaneous labour may represent a local intrauterine progesterone withdrawal, which, together with other signals, would activate parturition pathways involving arachidonate and sphingosine metabolism.
Collapse
Affiliation(s)
- Katherine Alice Birchenall
- Department of Obstetrics and Gynaecology, St Michael's Hospital, Southwell Street, Bristol, BS2 8EG, UK; Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Gavin Iain Welsh
- Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Andrés López Bernal
- Department of Obstetrics and Gynaecology, St Michael's Hospital, Southwell Street, Bristol, BS2 8EG, UK; Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
21
|
Garrett AS, Means SA, Roesler MW, Miller KJW, Cheng LK, Clark AR. Modeling and experimental approaches for elucidating multi-scale uterine smooth muscle electro- and mechano-physiology: A review. Front Physiol 2022; 13:1017649. [PMID: 36277190 PMCID: PMC9585314 DOI: 10.3389/fphys.2022.1017649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
The uterus provides protection and nourishment (via its blood supply) to a developing fetus, and contracts to deliver the baby at an appropriate time, thereby having a critical contribution to the life of every human. However, despite this vital role, it is an under-investigated organ, and gaps remain in our understanding of how contractions are initiated or coordinated. The uterus is a smooth muscle organ that undergoes variations in its contractile function in response to hormonal fluctuations, the extreme instance of this being during pregnancy and labor. Researchers typically use various approaches to studying this organ, such as experiments on uterine muscle cells, tissue samples, or the intact organ, or the employment of mathematical models to simulate the electrical, mechanical and ionic activity. The complexity exhibited in the coordinated contractions of the uterus remains a challenge to understand, requiring coordinated solutions from different research fields. This review investigates differences in the underlying physiology between human and common animal models utilized in experiments, and the experimental interventions and computational models used to assess uterine function. We look to a future of hybrid experimental interventions and modeling techniques that could be employed to improve the understanding of the mechanisms enabling the healthy function of the uterus.
Collapse
Affiliation(s)
| | | | | | | | | | - Alys R. Clark
- Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
GABAergic and Glutamatergic Phenotypes of Neurons Expressing Calcium-Binding Proteins in the Preoptic Area of the Guinea Pig. Int J Mol Sci 2022; 23:ijms23147963. [PMID: 35887305 PMCID: PMC9320123 DOI: 10.3390/ijms23147963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
The mammalian preoptic area (POA) has large populations of calbindin (CB), calretinin (CR) and parvalbumin (PV) neurons, but phenotypes of these cells are unknown. Therefore, the question is whether neurons expressing CB, CR, and/or PV are GABAergic or glutamatergic. Double-immunofluorescence staining followed by epifluorescence and confocal microscopy was used to determine the coexpression patterns of CB, CR and PV expressing neurons with vesicular GABA transporters (VGAT) as specific markers of GABAergic neurons and vesicular glutamate transporters (VGLUT 2) as specific markers of glutamatergic neurons. The guinea pig was adopted as, like humans, it has a reproductive cycle with a true luteal phase and a long gestation period. The results demonstrated that in the guinea pig POA of both sexes, ~80% of CB+ and ~90% of CR+ neurons coexpress VGAT; however, one-fifth of CB+ neurons and one-third of CR+ cells coexpress VGLUT. About two-thirds of PV+ neurons express VGAT, and similar proportion of them coexpress VGLUT. Thus, many CB+, CR+ and PV+ neurons may be exclusively GABAergic (VGAT-expressing cells) or glutamatergic (VGLUT-expressing cells); however, at least a small fraction of CR+ cells and at least one-third of PV+ cells are likely neurons with a dual GABA/glutamate phenotype that may coexpress both transporters.
Collapse
|
23
|
Lee H, Patel V, Onushko M, Fang X, Chemtob S, Olson D. A Leukocyte Migration Assay Assists Understanding of Interleukin-1β-Induced Leukocyte Migration Into Preterm Mouse Uterus. Front Pharmacol 2022; 13:898008. [PMID: 35694257 PMCID: PMC9174527 DOI: 10.3389/fphar.2022.898008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/18/2022] [Indexed: 11/24/2022] Open
Abstract
Neutrophils and other leukocytes invade the mouse uterus at term birth, which is normal for activating the uterus for labor. To better understand the regulation of this migration at term and interleukin (IL)-1β—induced preterm birth, we developed a mouse leukocyte migration assay (mLMA) and used it with rytvela, an IL-1 receptor allosteric antagonist. The mLMA uses term peripheral blood leukocytes that migrate in a Boyden chamber in response to a chemoattractant. We tested several mouse uterine tissues after homogenization and sedimentation of debris for chemoattractant activity. The most active chemoattractant homogenate came from the mouse lower uterus on gestational day (GD) 18.5. Using flow cytometry, we demonstrated that 99% of the cells that migrate are neutrophils. IL-1β administered on GD 16 stimulated neutrophil migration and invasion into the uterus and the fetal brain along with preterm birth on GD 17. Preterm birth and the increased leukocyte invasion of the maternal uterus and fetal brain were all blocked by the co-administration of rytvela. To test where the site of IL-1β action might be, we examined the potency of lower uterine chemoattractant and the activation of leukocytes following IL-1β +/- rytvela administration. IL-1β did not increase lower uterus homogenate chemoattractant activity, but it significantly (p < 0.05) increased leukocyte activation as defined by cytokine and chemokine expression. Rytvela blocked this activation of leukocytes by IL-1β. We conclude that IL-1β stimulates preterm birth in mice by increasing leukocyte activation leading to increased uterine and fetal brain leukocyte invasion.
Collapse
Affiliation(s)
- Han Lee
- Olson Laboratory, Department of Physiology, University of Alberta, Edmonton, AB, Canada
| | - Vaishvi Patel
- Olson Laboratory, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Meghan Onushko
- Olson Laboratory, Faculty of Science, University of Alberta, Edmonton, AB, Canada
| | - Xin Fang
- Olson Laboratory, Department of Obstetrics/Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Sylvain Chemtob
- Chemtob Laboratory, Departments of Pediatrics and Ophthalmology/Pharmacology, University of Montreal, Montreal, QC, Canada
| | - David Olson
- Olson Laboratory, Department of Physiology, University of Alberta, Edmonton, AB, Canada
- Olson Laboratory, Department of Obstetrics/Gynecology, University of Alberta, Edmonton, AB, Canada
- Olson Laboratory, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- *Correspondence: David Olson,
| |
Collapse
|
24
|
Winters AD, Romero R, Greenberg JM, Galaz J, Shaffer ZD, Garcia-Flores V, Kracht DJ, Gomez-Lopez N, Theis KR. Does the Amniotic Fluid of Mice Contain a Viable Microbiota? Front Immunol 2022; 13:820366. [PMID: 35296083 PMCID: PMC8920496 DOI: 10.3389/fimmu.2022.820366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/03/2022] [Indexed: 11/13/2022] Open
Abstract
The existence of an amniotic fluid microbiota (i.e., a viable microbial community) in mammals is controversial. Its existence would require a fundamental reconsideration of fetal in utero exposure to and colonization by microorganisms and the role of intra-amniotic microorganisms in fetal immune development as well as in pregnancy outcomes. In this study, we determined whether the amniotic fluid of mice harbors a microbiota in late gestation. The profiles of the amniotic fluids of pups located proximally or distally to the cervix were characterized through quantitative real-time PCR, 16S rRNA gene sequencing, and culture (N = 21 dams). These profiles were compared to those of technical controls for bacterial and DNA contamination. The load of 16S rRNA genes in the amniotic fluid exceeded that in controls. Additionally, the 16S rRNA gene profiles of the amniotic fluid differed from those of controls, with Corynebacterium tuberculostearicum being differentially more abundant in amniotic fluid profiles; however, this bacterium was not cultured from amniotic fluid. Of the 42 attempted bacterial cultures of amniotic fluids, only one yielded bacterial growth – Lactobacillus murinus. The 16S rRNA gene of this common murine-associated bacterium was not detected in any amniotic fluid sample, suggesting it did not originate from the amniotic fluid. No differences in the 16S rRNA gene load, 16S rRNA gene profile, or bacterial culture were observed between the amniotic fluids located Proximally and distally to the cervix. Collectively, these data indicate that, although there is a modest DNA signal of bacteria in murine amniotic fluid, there is no evidence that this signal represents a viable microbiota. While this means that amniotic fluid is not a source of microorganisms for in utero colonization in mice, it may nevertheless contribute to fetal exposure to microbial components. The developmental consequences of this observation warrant further investigation.
Collapse
Affiliation(s)
- Andrew D. Winters
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, United States
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, United States
- Detroit Medical Center, Detroit, MI, United States
| | - Jonathan M. Greenberg
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Zachary D. Shaffer
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, United States
- MD/PhD Combined Degree Program, Wayne State University School of Medicine, Detroit, MI, United States
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - David J. Kracht
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- *Correspondence: Kevin R. Theis, ; Nardhy Gomez-Lopez,
| | - Kevin R. Theis
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Detroit, MI, United States
- Perinatal Research Initiative in Maternal, Perinatal and Child Health, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, United States
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, MI, United States
- *Correspondence: Kevin R. Theis, ; Nardhy Gomez-Lopez,
| |
Collapse
|
25
|
Coker SJ, Smith-Díaz CC, Dyson RM, Vissers MCM, Berry MJ. The Epigenetic Role of Vitamin C in Neurodevelopment. Int J Mol Sci 2022; 23:ijms23031208. [PMID: 35163133 PMCID: PMC8836017 DOI: 10.3390/ijms23031208] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
The maternal diet during pregnancy is a key determinant of offspring health. Early studies have linked poor maternal nutrition during gestation with a propensity for the development of chronic conditions in offspring. These conditions include cardiovascular disease, type 2 diabetes and even compromised mental health. While multiple factors may contribute to these outcomes, disturbed epigenetic programming during early development is one potential biological mechanism. The epigenome is programmed primarily in utero, and during this time, the developing fetus is highly susceptible to environmental factors such as nutritional insults. During neurodevelopment, epigenetic programming coordinates the formation of primitive central nervous system structures, neurogenesis, and neuroplasticity. Dysregulated epigenetic programming has been implicated in the aetiology of several neurodevelopmental disorders such as Tatton-Brown-Rahman syndrome. Accordingly, there is great interest in determining how maternal nutrient availability in pregnancy might affect the epigenetic status of offspring, and how such influences may present phenotypically. In recent years, a number of epigenetic enzymes that are active during embryonic development have been found to require vitamin C as a cofactor. These enzymes include the ten-eleven translocation methylcytosine dioxygenases (TETs) and the Jumonji C domain-containing histone lysine demethylases that catalyse the oxidative removal of methyl groups on cytosines and histone lysine residues, respectively. These enzymes are integral to epigenetic regulation and have fundamental roles in cellular differentiation, the maintenance of pluripotency and development. The dependence of these enzymes on vitamin C for optimal catalytic activity illustrates a potentially critical contribution of the nutrient during mammalian development. These insights also highlight a potential risk associated with vitamin C insufficiency during pregnancy. The link between vitamin C insufficiency and development is particularly apparent in the context of neurodevelopment and high vitamin C concentrations in the brain are indicative of important functional requirements in this organ. Accordingly, this review considers the evidence for the potential impact of maternal vitamin C status on neurodevelopmental epigenetics.
Collapse
Affiliation(s)
- Sharna J. Coker
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Carlos C. Smith-Díaz
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - Rebecca M. Dyson
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
| | - Margreet C. M. Vissers
- Centre for Free Radical Research, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
- Correspondence: (M.C.M.V.); (M.J.B.)
| | - Mary J. Berry
- Perinatal & Developmental Physiology Group, Department of Paediatrics & Child Health, University of Otago, Wellington 6242, New Zealand; (S.J.C.); (R.M.D.)
- Correspondence: (M.C.M.V.); (M.J.B.)
| |
Collapse
|
26
|
Leimert KB, Xu W, Princ MM, Chemtob S, Olson DM. Inflammatory Amplification: A Central Tenet of Uterine Transition for Labor. Front Cell Infect Microbiol 2021; 11:660983. [PMID: 34490133 PMCID: PMC8417473 DOI: 10.3389/fcimb.2021.660983] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/30/2021] [Indexed: 11/23/2022] Open
Abstract
In preparation for delivery, the uterus transitions from actively maintaining quiescence during pregnancy to an active parturient state. This transition occurs as a result of the accumulation of pro-inflammatory signals which are amplified by positive feedback interactions involving paracrine and autocrine signaling at the level of each intrauterine cell and tissue. The amplification events occur in parallel until they reach a certain threshold, ‘tipping the scale’ and contributing to processes of uterine activation and functional progesterone withdrawal. The described signaling interactions all occur upstream from the presentation of clinical labor symptoms. In this review, we will: 1) describe the different physiological processes involved in uterine transition for each intrauterine tissue; 2) compare and contrast the current models of labor initiation; 3) introduce innovative models for measuring paracrine inflammatory interactions; and 4) discuss the therapeutic value in identifying and targeting key players in this crucial event for preterm birth.
Collapse
Affiliation(s)
- Kelycia B Leimert
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Wendy Xu
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Magdalena M Princ
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Montreal, QC, Canada
| | - David M Olson
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
27
|
High Mobility Group Box 1 in Pig Amniotic Membrane Experimentally Infected with E. coli O55. Biomolecules 2021; 11:biom11081146. [PMID: 34439812 PMCID: PMC8393629 DOI: 10.3390/biom11081146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/15/2022] Open
Abstract
Intra-amniotic infections (IAI) are one of the reasons for preterm birth. High mobility group box 1 (HMGB1) is a nuclear protein with various physiological functions, including tissue healing. Its excessive extracellular release potentiates inflammatory reaction and can revert its action from beneficial to detrimental. We infected the amniotic fluid of a pig on the 80th day of gestation with 1 × 104 colony forming units (CFUs) of E. coli O55 for 10 h, and evaluated the appearance of HMGB1, receptor for glycation endproducts (RAGE), and Toll-like receptor (TLR) 4 in the amniotic membrane and fluid. Sham-infected amniotic fluid served as a control. The expression and release of HMGB1 were evaluated by Real-Time PCR, immunofluorescence, immunohistochemistry, and ELISA. The infection downregulated HMGB1 mRNA expression in the amniotic membrane, changed the distribution of HMGB1 protein in the amniotic membrane, and increased its level in amniotic fluid. All RAGE mRNA, protein expression in the amniotic membrane, and soluble RAGE level in the amniotic fluid were downregulated. TLR4 mRNA and protein expression and soluble TLR4 were all upregulated. HMGB1 is a potential target for therapy to suppress the exaggerated inflammatory response. This controlled expression and release can, in some cases, prevent the preterm birth of vulnerable infants. Studies on suitable animal models can contribute to the development of appropriate therapy.
Collapse
|
28
|
Carter AM. Unique Aspects of Human Placentation. Int J Mol Sci 2021; 22:8099. [PMID: 34360862 PMCID: PMC8347521 DOI: 10.3390/ijms22158099] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/22/2022] Open
Abstract
Human placentation differs from that of other mammals. A suite of characteristics is shared with haplorrhine primates, including early development of the embryonic membranes and placental hormones such as chorionic gonadotrophin and placental lactogen. A comparable architecture of the intervillous space is found only in Old World monkeys and apes. The routes of trophoblast invasion and the precise role of extravillous trophoblast in uterine artery transformation is similar in chimpanzee and gorilla. Extended parental care is shared with the great apes, and though human babies are rather helpless at birth, they are well developed (precocial) in other respects. Primates and rodents last shared a common ancestor in the Cretaceous period, and their placentation has evolved independently for some 80 million years. This is reflected in many aspects of their placentation. Some apparent resemblances such as interstitial implantation and placental lactogens are the result of convergent evolution. For rodent models such as the mouse, the differences are compounded by short gestations leading to the delivery of poorly developed (altricial) young.
Collapse
Affiliation(s)
- Anthony M Carter
- Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, DK-5230 Odense, Denmark
| |
Collapse
|
29
|
Khader N, Shchuka VM, Shynlova O, Mitchell JA. Transcriptional control of parturition: insights from gene regulation studies in the myometrium. Mol Hum Reprod 2021; 27:gaab024. [PMID: 33823545 PMCID: PMC8126590 DOI: 10.1093/molehr/gaab024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
The onset of labour is a culmination of a series of highly coordinated and preparatory physiological events that take place throughout the gestational period. In order to produce the associated contractions needed for foetal delivery, smooth muscle cells in the muscular layer of the uterus (i.e. myometrium) undergo a transition from quiescent to contractile phenotypes. Here, we present the current understanding of the roles transcription factors play in critical labour-associated gene expression changes as part of the molecular mechanistic basis for this transition. Consideration is given to both transcription factors that have been well-studied in a myometrial context, i.e. activator protein 1, progesterone receptors, oestrogen receptors, and nuclear factor kappa B, as well as additional transcription factors whose gestational event-driving contributions have been demonstrated more recently. These transcription factors may form pregnancy- and labour-associated transcriptional regulatory networks in the myometrium to modulate the timing of labour onset. A more thorough understanding of the transcription factor-mediated, labour-promoting regulatory pathways holds promise for the development of new therapeutic treatments that can be used for the prevention of preterm labour in at-risk women.
Collapse
Affiliation(s)
- Nawrah Khader
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Virlana M Shchuka
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Oksana Shynlova
- Lunenfeld Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Obstetrics & Gynaecology, University of Toronto, ON, Canada
| | - Jennifer A Mitchell
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
30
|
Miller A, Jentz E, Duncan C, Merriman D. Progestogen metabolites for use in pregnancy monitoring of 13-lined ground squirrels ( Ictidomys tridecemlineatus). REPRODUCTION AND FERTILITY 2021; 2:81-88. [PMID: 35128444 PMCID: PMC8812425 DOI: 10.1530/raf-20-0071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/18/2021] [Indexed: 01/31/2023] Open
Abstract
13-lined ground squirrels (TLGS; Ictidomys tridecemlineatus) are small, omnivorous, fossorial, hibernating sciurids. TLGS are seasonal induced ovulators, with a ~28-day gestation period. The main goal of this study was to ascertain whether enzyme-linked immunosorbent assay (ELISA) of TLGS fecal samples can be used to non-invasively detect pregnancy. Competitive ELISAs for progestogen metabolites were conducted on feces collected from a group of (n =13) females. Feces were collected thrice weekly during the breeding season and frozen for subsequent analysis. Competitive ELISAs were run using progesterone kits ), setting data against seven different time-points between hibernation, emergence, and litter birthdate. Eleven females produced litters. ELISA data from the (n = 2) non-pregnant females demonstrated no rise in progestogen metabolites at any point over 28 days. In contrast, data from the (n = 11) pregnant females all demonstrated a pronounced rise in progestogen metabolites, with most animals displaying progesterone withdrawal in the final week of gestation. A >20-fold rise in progestogen metabolite was observed halfway through gestation (P < 005). Analysis on litter size and progestogen metabolite concentration showed no significant correlation (r2 = -0.615). Initial correlation analysis done on sex ratio of litters vs progestogen metabolites showed no significant effect of progesterone on sex ratios (males: r2 = -0.772, females: r2 = 0.375). This work demonstrated that TLGS also undergo progesterone withdrawal about a week before parturition. We have ascertained that a commercially available progesterone assay kit can detect a significant elevation in progestogen metabolites in this species about halfway through gestation. LAY SUMMARY This research was conducted to discover whether pregnancy prediction is possible in female 13-lined ground squirrels (TLGS; a small hibernating ground squirrel named for their number of stripes). Pregnancy status in this species, we postulated, could be anticipated by generating profiles for individuals via a non-invasive technique known as fecal endocrine hormone profiling. Fecal samples were collected from 13 females thrice weekly for 4 weeks post-hibernation in the breeding season of 2016. Fecal samples were then processed and run through an assay known as an ELISA giving concentrations of hormone metabolites excreted through feces. We then set these samples against time points to develop a profile for each female. We have ascertained that elevated progesterone (potential pregnancy) can be detected by a commercially available assay kit. Understanding hormone patterns in animals gives researchers a better idea of best husbandry practices, including breeding in managed care.
Collapse
Affiliation(s)
- Amy Miller
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, USA,Correspondence should be addressed to A Miller:
| | - Elainna Jentz
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, USA
| | - Cassandra Duncan
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, USA
| | - Dana Merriman
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, USA
| |
Collapse
|
31
|
Nold C, Esteves K, Jensen T, Vella AT. Granulocyte-macrophage colony-stimulating factor initiates amniotic membrane rupture and preterm birth in a mouse model. Am J Reprod Immunol 2021; 86:e13424. [PMID: 33772943 DOI: 10.1111/aji.13424] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/05/2021] [Accepted: 03/23/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Preterm premature rupture of membranes is associated with 30% of all preterm births. The weakening of amniotic membranes is associated with an increase in matrix metallopeptidases (MMPs) along with a decrease in their inhibitors, tissue inhibitor metallopeptidases (TIMPs). Additionally, granulocyte-macrophage colony-stimulating factor (GM-CSF) has been shown to weaken fetal membranes in-vitro. We hypothesize pregnant mice treated with GM-CSF lead to increased MMPs:TIMPs resulting in membrane rupture and preterm birth. STUDY DESIGN Pregnant CD-1 mice on gestational day 17 received either an intrauterine injection of GM-CSF or vehicle control. A second series of mice were administered an intrauterine injection of Lipopolysaccharide along with either anti-mouse GM-CSF or control antibody. Mice were evaluated for rupture of membranes and/or preterm birth and the uterus, amniotic fluid, and serum were collected for analysis. RESULTS 87.5% of GM-CSF mice exhibited evidence of membrane rupture or preterm birth, compared with 0% in control mice (p < .001). Treatment with GM-CSF decreased the expression of TNFα (p < .05) while increasing the ratio of MMP2:TIMP1 (p < .05), MMP2:TIMP2 (p < .05), MMP2:TIMP3 (p < .001), MMP9:TIMP1 (p < .01), MMP9:TIMP2 (p < .05), MMP9:TIMP3 (p < .001), and MMP10:TIMP1 (p < .05). Mice treated with LPS and the GM-CSF antibody resulted in a decrease in the ratio of MMP2:TIMP1 (p < .0001) compared with controls. CONCLUSION These studies demonstrate GM-CSF will result in membrane rupture and preterm birth by increasing the ratio MMPs:TIMPs in our animal model. By increasing our understanding of the molecular pathways associated with GM-CSF, we may be able to develop future therapies to prevent preterm birth and reduce neonatal morbidity.
Collapse
Affiliation(s)
- Christopher Nold
- Department of Women's Health, Hartford Hospital, Hartford, CT, USA.,Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Kristyn Esteves
- Department of Obstetrics and Gynecology, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Todd Jensen
- Department of Pediatrics, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Anthony T Vella
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, USA
| |
Collapse
|
32
|
Malik M, Roh M, England SK. Uterine contractions in rodent models and humans. Acta Physiol (Oxf) 2021; 231:e13607. [PMID: 33337577 PMCID: PMC8047897 DOI: 10.1111/apha.13607] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022]
Abstract
Aberrant uterine contractions can lead to preterm birth and other labour complications and are a significant cause of maternal morbidity and mortality. To investigate the mechanisms underlying dysfunctional uterine contractions, researchers have used experimentally tractable small animal models. However, biological differences between humans and rodents change how researchers select their animal model and interpret their results. Here, we provide a general review of studies of uterine excitation and contractions in mice, rats, guinea pigs, and humans, in an effort to introduce new researchers to the field and help in the design and interpretation of experiments in rodent models.
Collapse
Affiliation(s)
- Manasi Malik
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Michelle Roh
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Sarah K. England
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
33
|
Alan E, Liman N. The distribution and immunolocalization of fibroblast growth factors (FGFs) in the rat oviduct during early pregnancy and the post-partum period. Anat Histol Embryol 2021; 50:645-657. [PMID: 33772852 DOI: 10.1111/ahe.12672] [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: 11/26/2020] [Revised: 02/28/2021] [Accepted: 03/15/2021] [Indexed: 11/29/2022]
Abstract
The mammalian oviduct provides a favourable environment for several reproductive processes, including ovum transport, sperm capacitation, fertilization and pre-implantation embryonic development. This environment is regulated by cyclic ovarian steroids, that is oestrogen, and growth factors. Fibroblast growth factors (FGFs) regulate the differentiation and growth of various cell types in the female genital tract. This study aimed to determine the localization of FGF1, FGF2, FGF receptor 1 (FGFR1) and 2 (FGFR2) in the rat oviduct, by immunohistochemistry, on day 5 of pregnancy and post-partum days 1, 3 and 5, and to demonstrate the possible functions of these proteins during early pregnancy and the post-partum period. On all examination days, cytoplasmic and nuclear FGF1 immunoreactivity was detected in the epithelium lining the infundibulum, ampulla and isthmus of the oviduct. Immunoreactivity was much stronger in the basal bodies of the cilia on the epithelium lining the infundibulum and ampulla. FGF1 immunoreactivity was also detected in stromal cells, myocytes and endothelial cells. Cytoplasmic FGF2 immunoreactivity was observed in the tunica muscularis, vascular myocytes and endothelial cells. While strong cytoplasmic FGF2 immunoreactions were observed in the stromal cells of the lamina propria, the luminal epithelium, some stromal cells and smooth muscle cells displayed a rather weak FGFR1 and FGFR2 immunoreactivity. Immunoreaction intensity did not differ between the periods examined. This study shows that FGF1, FGF2, FGFR1 and FGFR2 are produced by rat oviduct cells during pregnancy and the post-partum period, and reproductive physiology is regulated not only by hormonal mechanisms, but also by growth factors.
Collapse
Affiliation(s)
- Emel Alan
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| | - Narin Liman
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Erciyes University, Kayseri, Turkey
| |
Collapse
|
34
|
Progesterone receptor isoform B regulates the Oxtr- Plcl2- Trpc3 pathway to suppress uterine contractility. Proc Natl Acad Sci U S A 2021; 118:2011643118. [PMID: 33707208 DOI: 10.1073/pnas.2011643118] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Uterine contractile dysfunction leads to pregnancy complications such as preterm birth and labor dystocia. In humans, it is hypothesized that progesterone receptor isoform PGR-B promotes a relaxed state of the myometrium, and PGR-A facilitates uterine contraction. This hypothesis was tested in vivo using transgenic mouse models that overexpress PGR-A or PGR-B in smooth muscle cells. Elevated PGR-B abundance results in a marked increase in gestational length compared to control mice (21.1 versus 19.1 d respectively, P < 0.05). In both ex vivo and in vivo experiments, PGR-B overexpression leads to prolonged labor, a significant decrease in uterine contractility, and a high incidence of labor dystocia. Conversely, PGR-A overexpression leads to an increase in uterine contractility without a change in gestational length. Uterine RNA sequencing at midpregnancy identified 1,174 isoform-specific downstream targets and 424 genes that are commonly regulated by both PGR isoforms. Gene signature analyses further reveal PGR-B for muscle relaxation and PGR-A being proinflammatory. Elevated PGR-B abundance reduces Oxtr and Trpc3 and increases Plcl2 expression, which manifests a genetic profile of compromised oxytocin signaling. Functionally, both endogenous PLCL2 and its paralog PLCL1 can attenuate uterine muscle cell contraction in a CRISPRa-based assay system. These findings provide in vivo support that PGR isoform levels determine distinct transcriptomic landscapes and pathways in myometrial function and labor, which may help further the understanding of abnormal uterine function in the clinical setting.
Collapse
|
35
|
Maxey AP, McCain ML. Tools, techniques, and future opportunities for characterizing the mechanobiology of uterine myometrium. Exp Biol Med (Maywood) 2021; 246:1025-1035. [PMID: 33554648 DOI: 10.1177/1535370221989259] [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] [Indexed: 11/15/2022] Open
Abstract
The myometrium is the smooth muscle layer of the uterus that generates the contractions that drive processes such as menstruation and childbirth. Aberrant contractions of the myometrium can result in preterm birth, insufficient progression of labor, or other difficulties that can lead to maternal or fetal complications or even death. To investigate the underlying mechanisms of these conditions, the most common model systems have conventionally been animal models and human tissue strips, which have limitations mostly related to relevance and scalability, respectively. Myometrial smooth muscle cells have also been isolated from patient biopsies and cultured in vitro as a more controlled experimental system. However, in vitro approaches have focused primarily on measuring the effects of biochemical stimuli and neglected biomechanical stimuli, despite the extensive evidence indicating that remodeling of tissue rigidity or excessive strain is associated with uterine disorders. In this review, we first describe the existing approaches for modeling human myometrium with animal models and human tissue strips and compare their advantages and disadvantages. Next, we introduce existing in vitro techniques and assays for assessing contractility and summarize their applications in elucidating the role of biochemical or biomechanical stimuli on human myometrium. Finally, we conclude by proposing the translation of "organ on chip" approaches to myometrial smooth muscle cells as new paradigms for establishing their fundamental mechanobiology and to serve as next-generation platforms for drug development.
Collapse
Affiliation(s)
- Antonina P Maxey
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Megan L McCain
- Laboratory for Living Systems Engineering, Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
36
|
Hyaluronidase Impairs Neutrophil Function and Promotes Group B Streptococcus Invasion and Preterm Labor in Nonhuman Primates. mBio 2021; 12:mBio.03115-20. [PMID: 33402537 PMCID: PMC8545101 DOI: 10.1128/mbio.03115-20] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Invasive bacterial infections during pregnancy are a major risk factor for preterm birth, stillbirth, and fetal injury. Group B streptococci (GBS) are Gram-positive bacteria that asymptomatically colonize the lower genital tract but infect the amniotic fluid and induce preterm birth or stillbirth. Experimental models that closely emulate human pregnancy are pivotal for the development of successful strategies to prevent these adverse pregnancy outcomes. Using a unique nonhuman primate model that mimics human pregnancy and informs temporal events surrounding amniotic cavity invasion and preterm labor, we show that the animals inoculated with hyaluronidase (HylB)-expressing GBS consistently exhibited microbial invasion into the amniotic cavity, fetal bacteremia, and preterm labor. Although delayed cytokine responses were observed at the maternal-fetal interface, increased prostaglandin and matrix metalloproteinase levels in these animals likely mediated preterm labor. HylB-proficient GBS dampened reactive oxygen species production and exhibited increased resistance to neutrophils compared to an isogenic mutant. Together, these findings demonstrate how a bacterial enzyme promotes GBS amniotic cavity invasion and preterm labor in a model that closely resembles human pregnancy.
Collapse
|
37
|
Block LN, Bowman BD, Schmidt JK, Keding LT, Stanic AK, Golos TG. The promise of placental extracellular vesicles: models and challenges for diagnosing placental dysfunction in utero†. Biol Reprod 2021; 104:27-57. [PMID: 32856695 PMCID: PMC7786267 DOI: 10.1093/biolre/ioaa152] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/04/2020] [Accepted: 08/26/2020] [Indexed: 12/14/2022] Open
Abstract
Monitoring the health of a pregnancy is of utmost importance to both the fetus and the mother. The diagnosis of pregnancy complications typically occurs after the manifestation of symptoms, and limited preventative measures or effective treatments are available. Traditionally, pregnancy health is evaluated by analyzing maternal serum hormone levels, genetic testing, ultrasonographic imaging, and monitoring maternal symptoms. However, researchers have reported a difference in extracellular vesicle (EV) quantity and cargo between healthy and at-risk pregnancies. Thus, placental EVs (PEVs) may help to understand normal and aberrant placental development, monitor pregnancy health in terms of developing placental pathologies, and assess the impact of environmental influences, such as infection, on pregnancy. The diagnostic potential of PEVs could allow for earlier detection of pregnancy complications via noninvasive sampling and frequent monitoring. Understanding how PEVs serve as a means of communication with maternal cells and recognizing their potential utility as a readout of placental health have sparked a growing interest in basic and translational research. However, to date, PEV research with animal models lags behind human studies. The strength of animal pregnancy models is that they can be used to assess placental pathologies in conjunction with isolation of PEVs from fluid samples at different time points throughout gestation. Assessing PEV cargo in animals within normal and complicated pregnancies will accelerate the translation of PEV analysis into the clinic for potential use in prognostics. We propose that appropriate animal models of human pregnancy complications must be established in the PEV field.
Collapse
Affiliation(s)
- Lindsey N Block
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Brittany D Bowman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Jenna Kropp Schmidt
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Logan T Keding
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Aleksandar K Stanic
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus G Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
38
|
Rokas A, Mesiano S, Tamam O, LaBella A, Zhang G, Muglia L. Developing a theoretical evolutionary framework to solve the mystery of parturition initiation. eLife 2020; 9:e58343. [PMID: 33380346 PMCID: PMC7775106 DOI: 10.7554/elife.58343] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 12/08/2020] [Indexed: 11/13/2022] Open
Abstract
Eutherian mammals have characteristic lengths of gestation that are key for reproductive success, but relatively little is known about the processes that determine the timing of parturition, the process of birth, and how they are coordinated with fetal developmental programs. This issue remains one of biology's great unsolved mysteries and has significant clinical relevance because preterm birth is the leading cause of infant and under 5 year old child mortality worldwide. Here, we consider the evolutionary influences and potential signaling mechanisms that maintain or end pregnancy in eutherian mammals and use this knowledge to formulate general theoretical evolutionary models. These models can be tested through evolutionary species comparisons, studies of experimental manipulation of gestation period and birth timing, and human clinical studies. Understanding how gestation time and parturition are determined will shed light on this fundamental biological process and improve human health through the development of therapies to prevent preterm birth.
Collapse
Affiliation(s)
- Antonis Rokas
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Sam Mesiano
- Department of Reproductive Biology, Case Western Reserve University and Department of Obstetrics and Gynecology, University Hospitals of Cleveland, Cleveland, United States
| | - Ortal Tamam
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben Gurion University, Beer Sheva, Israel
| | - Abigail LaBella
- Department of Biological Sciences, Vanderbilt University, Nashville, United States
| | - Ge Zhang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics. University of Cincinnati College of Medicine, Cincinnati, United States
| | - Louis Muglia
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics. University of Cincinnati College of Medicine, Cincinnati, United States
- Burroughs Wellcome Fund, Research Triangle Park, Durham, United States
| |
Collapse
|
39
|
Valdés G, Acuña S, Schneider D, Ortíz R, Padilla O. Bradykinin Exerts Independent Effects on Trophoblast Invasion and Blood Pressure in Pregnant Guinea Pigs. Reprod Sci 2020; 27:1648-1655. [PMID: 32430711 DOI: 10.1007/s43032-020-00195-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The pleiotropic kininogen-kallikrein-kinin system is upregulated in pregnancy and localizes in the uteroplacental unit. To identify the systemic and local participation of the bradykinin type 2 receptor (B2R), this was antagonized by Bradyzide (BDZ) during 2 periods: from days 20 to 34 and from days 20 to 60 in pregnant guinea pigs. METHODS Pregnant guinea pigs received subcutaneous infusions of saline or BDZ from gestational day 20 until sacrifice on day 34 (Short B2R Antagonism [SH-B2RA]) or on day 60 (Prolonged B2R Antagonism [PR-B2RA]). In SH-BDZA, systolic blood pressure was determined on day 34, while in PR-BDZA it was measured preconceptionally, at days 40 and 60. On gestational day 60, plasma creatinine, uricemia, proteinuria, fetal, placental and maternal kidney weight, and the extent of trophoblast invasion were evaluated. RESULTS The SH-B2RA increased systolic blood pressure on day 34 and reduced trophoblast myometrial invasion, spiral artery remodeling, and placental sufficiency. The PR-B2RA suppressed the normal blood pressure fall observed on days 40 and 60; vascular transformation, placental efficiency, urinary protein, serum creatinine, and uric acid did not differ between the groups. The proportion of all studied mothers with lost fetuses was greater under BDZ infusion than in controls. CONCLUSION The increased systolic blood pressure and transient reduction in trophoblast invasion and fetal/placental weight in the SH-B2R blockade and the isolated impact on blood pressure in the PR-B2R blockade indicate that bradykinin independently modulates systemic hemodynamics and the uteroplacental unit through cognate vascular and local B2R receptors.
Collapse
Affiliation(s)
- Gloria Valdés
- Centro de Investigaciones Médicas, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile.
- Departamento de Nefrología, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile.
| | - Stephanie Acuña
- Centro de Investigaciones Médicas, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile
| | - Daniela Schneider
- Centro de Investigaciones Médicas, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile
| | - Rita Ortíz
- Centro de Investigaciones Médicas, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile
| | - Oslando Padilla
- Departamento de Salud Pública, Facultad de Medicina, Pontificia Universidad Católica, Santiago, Chile
| |
Collapse
|
40
|
Qiu W, Hodges TE, Clark EL, Blankers SA, Galea LAM. Perinatal depression: Heterogeneity of disease and in animal models. Front Neuroendocrinol 2020; 59:100854. [PMID: 32750403 DOI: 10.1016/j.yfrne.2020.100854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
Perinatal depression (PND) can have either an antepartum or postpartum onset. Although the greatest risk factor for PND is previous depression history,de novoPND occurs with the majority of cases occurring in the postpartum. Timing of depression can impact etiology, prognosis, and response to treatment. Thus, it is crucial to study the impact of the heterogeneity of PND for better health outcomes. In this review, we outline the differences between antepartum and postpartum depression onset of PND. We discuss maternal physiological changes that differ between pregnancy and postpartum and how these may differentially impact depression susceptibility. We highlight changes in the maternal steroid and peptide hormone levels, immune signalling, serotonergic tone, metabolic factors, brain morphology, and the gut microbiome. Finally, we argue that studying the heterogeneity of PND in clinical and preclinical models can lead to improved knowledge of disease etiopathology and treatment outcomes.
Collapse
Affiliation(s)
- Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Travis E Hodges
- Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Emily L Clark
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
41
|
Çintesun E, Aydoğdu M, Akar S, Çelik Ç. Is striae gravidarum a sign of spontaneous premature birth? J Matern Fetal Neonatal Med 2020; 35:3467-3472. [PMID: 32957847 DOI: 10.1080/14767058.2020.1821642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND This study aimed to investigate the association of striae gravidarum (SG) and preterm delivery. MATERIAL AND METHODS This prospective cross-sectional study was performed between November 2018 and February 2019. In addition to demographic variables, SG score of the abdomen, presence of striae on breasts, arms, hips, and thighs were recorded. Davey's scoring system was used to define the severity of SG in the abdomen by the same investigators. Patients with cervical dilatation of 6 cm or more were included in the preterm labor group. RESULTS A total of 292 women were included in the study. Of these, 176 (60.3%) had no SG, 20 (6.8%) had mild SG, and 96 (32.9%) had severe SG. Davey's score was lower in the group of patients with preterm birth than in the term birth groups (p = .002). SG in the breasts was more common in the preterm labor group than in the term birth group (p = .007). Also, the presence of SG in the legs was less common in the preterm labor group than in the term birth group (p < .001). In a logistic regression model, stria in the breasts revealed most significant in preterm delivery. CONCLUSIONS No difference was found in the pregnancy length in gestational weeks among groups of different SG severity. The Davey's score and the presence of striae in the legs and breasts were found different between the preterm and term birth groups, and the term birth subgroups.
Collapse
Affiliation(s)
- Ersin Çintesun
- Department of Obstetrics and Gynecology, Selçuk University Medicine Faculty, Selçuklu, Konya, Turkey
| | - Meltem Aydoğdu
- Department of Obstetrics and Gynecology, Selçuk University Medicine Faculty, Selçuklu, Konya, Turkey
| | - Serra Akar
- Department of Obstetrics and Gynecology, Selçuk University Medicine Faculty, Selçuklu, Konya, Turkey
| | - Çetin Çelik
- Department of Obstetrics and Gynecology, Selçuk University Medicine Faculty, Selçuklu, Konya, Turkey
| |
Collapse
|
42
|
Clusters of Tolerogenic B Cells Feature in the Dynamic Immunological Landscape of the Pregnant Uterus. Cell Rep 2020; 32:108204. [PMID: 32997982 DOI: 10.1016/j.celrep.2020.108204] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Well-timed interaction of correctly functioning maternal immune cells is essential to facilitate healthy placenta formation, because the uterine immune environment has to tolerate the semi-allogeneic fetus and allow adequate trophoblast invasion. Here, we assess the uterine immune signature before and during pregnancy. Extensive supervised and unsupervised flow cytometry clustering strategies not only show a general increase in immune memory throughout pregnancy but also reveal the continuous presence of B cells. Contrary to the belief that B cells are merely a consequence of uterine pathology, decidual B cells produce IL-10 and are found to be localized in clusters, together with Foxp3pos T cells. Our findings therefore suggest a role for B cells in healthy pregnancy.
Collapse
|
43
|
Palomares KT, Parobchak N, Ithier MC, Aleksunes LM, Castaño PM, So M, Faro R, Heller D, Wang B, Rosen T. Fetal Exosomal Platelet-activating Factor Triggers Functional Progesterone Withdrawal in Human Placenta. Reprod Sci 2020; 28:252-262. [PMID: 32780361 DOI: 10.1007/s43032-020-00283-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 07/31/2020] [Indexed: 11/27/2022]
Abstract
In most mammals, labor is heralded by the withdrawal of progesterone. In humans, circulating progesterone levels increase as gestation advances while placental expression of progesterone receptor A (PR-A) declines. As a result of PR-A downregulation, the non-canonical NF-κB pathway is activated, an event implicated in triggering labor. Here, we sought to identify fetal-derived mediator(s) that represses placental PR-A in human placenta leading to activation of pro-labor signaling. Lipidomic profiling demonstrated enrichment of platelet-activating factor (PAF) in exosomes originating from the human fetus. Exposure of primary cytotrophoblasts to fetal exosomes from term pregnancies reduced PR-A expression by > 50%, and PAF also reduced PR-A message levels in a dose-dependent manner. Notably, fetal exosomes from preterm pregnancies had lower PAF levels and no effect on PR-A expression. Synthetic PAF-induced DNA methylation increases by 20% at the PR-A promoter, leading to recruitment of corepressors and downregulation of PR-A in cytotrophoblast. Furthermore, suppression of PR-A by PAF-stimulated expression of the pro-labor genes, corticotropin-releasing hormone (CRH) and cyclooxygenase-2 (COX-2), which was reversed by disruption of the DNA methyltransferases 3B and 3L. Taken together, PAF represents a novel fetal-derived candidate for initiation of labor by stimulating methylation and repression of PR-A and activating pro-labor signaling in trophoblast.
Collapse
Affiliation(s)
- Kristy T Palomares
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
- Department of Obstetrics and Gynecology, Saint Peter's University Hospital, New Brunswick, NJ, 08901, USA
| | - Nataliya Parobchak
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Mayra Cruz Ithier
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, 08854, USA
| | - Paula M Castaño
- Department of Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Melody So
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
| | - Revital Faro
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA
- Department of Obstetrics and Gynecology, Saint Peter's University Hospital, New Brunswick, NJ, 08901, USA
| | - Debra Heller
- Department of Pathology and Laboratory Medicine, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Bingbing Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| | - Todd Rosen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Division of Maternal-Fetal Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, 08901, USA.
| |
Collapse
|
44
|
Wen L, Li R, Wang J, Yi J. The reproductive stress hypothesis. Reproduction 2020; 158:R209-R218. [PMID: 31677601 PMCID: PMC6892456 DOI: 10.1530/rep-18-0592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 07/23/2019] [Indexed: 12/16/2022]
Abstract
In this paper, we propose the reproductive stress hypothesis that describes the pregnant females response to reproductive events based upon the activation of the hypothalamic–pituitary–adrenal axis and sympathetic adrenomedullary system. The main components of the reproductive stress hypothesis can be summarized as follows: (1) events unique to reproduction including empathema, pregnancy, parturition and lactation cause non-specific responses in females, called active reproductive stress; (2) the fetus is a special stressor for pregnant females where endocrine hormones, including corticotropin-releasing hormones and fetal glucocorticoids secreted by the fetus and placenta, enter the maternal circulatory system, leading to another stress response referred to as passive reproductive stress and (3) response to uterine tension and intrauterine infection is the third type of stress, called fetal intrauterine stress. Appropriate reproductive stress is a crucial prerequisite in normal reproductive processes. By contrast, excessive or inappropriate reproductive stress may result in dysfunctions of the reproductive system, such as compromised immune function, leading to susceptibility to disease. The novel insights of the reproductive stress hypothesis have important implications for deciphering the pathogenesis of certain diseases in pregnant animals, including humans, which in turn may be applied to preventing and treating their occurrence.
Collapse
Affiliation(s)
- Lixin Wen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, People's Republic of China.,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, People's Republic of China
| | - Rongfang Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, People's Republic of China.,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, People's Republic of China
| | - Ji Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, People's Republic of China.,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, People's Republic of China
| | - Jine Yi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Hunan Agricultural University, Changsha, People's Republic of China.,Hunan Collaborative Innovation Center of Animal Production Safety, Changsha, People's Republic of China
| |
Collapse
|
45
|
Gevi F, Meloni A, Mereu R, Lelli V, Chiodo A, Ragusa A, Timperio AM. Urine Metabolome during Parturition. Metabolites 2020; 10:metabo10070290. [PMID: 32708819 PMCID: PMC7407522 DOI: 10.3390/metabo10070290] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/30/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
In recent years, some studies have described metabolic changes during human childbirth labor. Metabolomics today is recognized as a powerful approach in a prenatal research context, since it can provide detailed information during pregnancy and it may enable the identification of biomarkers with potential diagnostic or predictive. This is an observational, longitudinal, prospective cohort study of a total of 51 serial urine samples from 15 healthy pregnant women, aged 29–40 years, which were collected before the onset of labor (out of labor, OL). In the same women, during labor (in labor or dilating phase, IL-DP). Samples were analyzed by hydrophilic interaction ultra-performance liquid chromatography coupled with mass spectrometry (HILIC-UPLC-MS), a highly sensitive, accurate, and unbiased approach. Metabolites were then subjected to multivariate statistical analysis and grouped by metabolic pathway. This method was used to identify the potential biomarkers. The top 20 most discriminative metabolites contributing to the complete separation of OL and IL-DP were identified. Urinary metabolites displaying the largest differences between OL and IL-DP belonged to steroid hormone, particularly conjugated estrogens and amino acids much of this difference is determined by the fetal contribution. In addition, our results highlighted the efficacy of using urine samples instead of more invasive techniques to evaluate the difference in metabolic analysis between OL and IL-DP.
Collapse
Affiliation(s)
- Federica Gevi
- Department of Biology and Ecology University of Tuscia, 01100 Viterbo, Italy; (F.G.); (V.L.)
| | - Alessandra Meloni
- Neonatal Department, Obstetrics and Gynecology Unit, Azienda Ospedaliera Universitaria (AOU), 09124 Cagliari, Italy; (A.M.); (R.M.); (A.C.)
| | - Rossella Mereu
- Neonatal Department, Obstetrics and Gynecology Unit, Azienda Ospedaliera Universitaria (AOU), 09124 Cagliari, Italy; (A.M.); (R.M.); (A.C.)
| | - Veronica Lelli
- Department of Biology and Ecology University of Tuscia, 01100 Viterbo, Italy; (F.G.); (V.L.)
| | - Antonella Chiodo
- Neonatal Department, Obstetrics and Gynecology Unit, Azienda Ospedaliera Universitaria (AOU), 09124 Cagliari, Italy; (A.M.); (R.M.); (A.C.)
| | - Antonio Ragusa
- Department of Obstetrics and Gynecology, Ospedale San Giovanni Calibita, Fatebenefratelli, Isola Tiberina, Via di Ponte Quattro Capi, 39, 00186 Roma, Italy;
| | - Anna Maria Timperio
- Department of Biology and Ecology University of Tuscia, 01100 Viterbo, Italy; (F.G.); (V.L.)
- Correspondence:
| |
Collapse
|
46
|
Wijaya JC, Khanabdali R, Georgiou HM, Kalionis B. Ageing in human parturition: impetus of the gestation clock in the decidua†. Biol Reprod 2020; 103:695-710. [PMID: 32591788 DOI: 10.1093/biolre/ioaa113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Despite sharing many common features, the relationship between ageing and parturition remains poorly understood. The decidua is a specialized lining of endometrial tissue, which develops in preparation for pregnancy. The structure and location of the decidua support its role as the physical scaffold for the growing embryo and placenta, and thus, it is vital to sustain pregnancy. Approaching term, the physical support properties of the decidua are naturally weakened to permit parturition. In this review, we hypothesize that the natural weakening of decidual tissue at parturition is promoted by the ageing process. Studies of the ageing-related functional and molecular changes in the decidua at parturition are reviewed and classified using hallmarks of ageing as the framework. The potential roles of decidual mesenchymal stem/stromal cell (DMSC) ageing in labor are also discussed because, although stem cell exhaustion is also a hallmark of ageing, its role in labor is not completely understood. In addition, the potential roles of extracellular vesicles secreted by DMSCs in labor, and their parturition-related miRNAs, are reviewed to gain further insight into this research area. In summary, the literature supports the notion that the decidua ages as the pregnancy progresses, and this may facilitate parturition, suggesting that ageing is the probable impetus of the gestational clocks in the decidua. This conceptual framework was developed to provide a better understanding of the natural ageing process of the decidua during parturition as well as to encourage future studies of the importance of healthy ageing for optimal pregnancy outcomes.
Collapse
Affiliation(s)
- Joan C Wijaya
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Ramin Khanabdali
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia.,Department of Process Development, Exopharm Limited, Melbourne, Victoria, Australia
| | - Harry M Georgiou
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Victoria, Australia.,University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Victoria, Australia
| |
Collapse
|
47
|
Yellon SM, Greaves E, Heuerman AC, Dobyns AE, Norman JE. Effects of macrophage depletion on characteristics of cervix remodeling and pregnancy in CD11b-dtr mice. Biol Reprod 2020; 100:1386-1394. [PMID: 30629144 DOI: 10.1093/biolre/ioz002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/30/2018] [Accepted: 01/07/2019] [Indexed: 12/24/2022] Open
Abstract
To test the hypothesis that macrophages are essential for remodeling the cervix in preparation for birth, pregnant homozygous CD11b-dtr mice were injected with diphtheria toxin (DT) on days 14 and 16 postbreeding. On day 15 postbreeding, macrophages (F4/80+) were depleted in cervix and kidney, but not in liver, ovary, or other non-reproductive tissues in DT-compared to saline-treated dtr mice or wild-type controls given DT or saline. Within 24 h of DT-treatment, the density of cell nuclei and macrophages declined in cervix stroma in dtr mice versus controls, but birefringence of collagen, as an indication of extracellular cross-linked structure, remained unchanged. Only in the cervix of DT-treated dtr mice was an apoptotic morphology evident in macrophages. DT-treatment did not alter the sparse presence or morphology of neutrophils. By day 18 postbreeding, macrophages repopulated the cervix in DT-treated dtr mice so that the numbers were comparable to that in controls. However, at term, evidence of fetal mortality without cervix ripening occurred in most dtr mice given DT-a possible consequence of treatment effects on placental function. These findings suggest that CD11b+ F4/80+ macrophages are important to sustain pregnancy and are required for processes that remodel the cervix in preparation for parturition.
Collapse
Affiliation(s)
- S M Yellon
- Longo Center for Perinatal Biology.,Division of Physiology, Departments of Basic Sciences, and Pediatrics, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - E Greaves
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, Scotland EH16 4TJ, United Kingdom
| | | | | | - J E Norman
- MRC Centre for Reproductive Health, Queens Medical Research Institute, University of Edinburgh, Edinburgh, Scotland EH16 4TJ, United Kingdom
| |
Collapse
|
48
|
Does early weaning shape future endocrine and metabolic disorders? Lessons from animal models. J Dev Orig Health Dis 2020; 11:441-451. [PMID: 32487270 DOI: 10.1017/s2040174420000410] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Obesity and its complications occur at alarming rates worldwide. Epidemiological data have associated perinatal conditions, such as malnutrition, with the development of some disorders, such as obesity, dyslipidemia, diabetes, and cardiovascular diseases, in childhood and adulthood. Exclusive breastfeeding has been associated with protection against long-term chronic diseases. However, in humans, the interruption of breastfeeding before the recommended period of 6 months is a common practice and can increase the risk of several metabolic disturbances. Nutritional and environmental changes within a critical window of development, such as pregnancy and breastfeeding, can induce permanent changes in metabolism through epigenetic mechanisms, leading to diseases later in life via a phenomenon known as programming or developmental plasticity. However, little is known regarding the underlying mechanisms by which precocious weaning can result in adipose tissue dysfunction and endocrine profile alterations. Here, the authors give a comprehensive report of the different animal models of early weaning and programming that can result in the development of metabolic syndrome. In rats, for example, pharmacological and nonpharmacological early weaning models are associated with the development of overweight and visceral fat accumulation, leptin and insulin resistance, and neuroendocrine and hepatic changes in adult progeny. Sex-related differences seem to influence this phenotype. Therefore, precocious weaning seems to be obesogenic for offspring. A better understanding of this condition seems essential to reducing the risk for diseases. Additionally, this knowledge can generate new insights into therapeutic strategies for obesity management, improving health outcomes.
Collapse
|
49
|
Inagaki M, Nishimura T, Nakanishi T, Shimada H, Noguchi S, Akanuma SI, Tachikawa M, Hosoya KI, Tamai I, Nakashima E, Tomi M. Contribution of Prostaglandin Transporter OATP2A1/SLCO2A1 to Placenta-to-Maternal Hormone Signaling and Labor Induction. iScience 2020; 23:101098. [PMID: 32408168 PMCID: PMC7225742 DOI: 10.1016/j.isci.2020.101098] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/22/2020] [Accepted: 04/21/2020] [Indexed: 01/09/2023] Open
Abstract
We evaluated the contribution of organic anion transporting polypeptide 2A1 (OATP2A1/SLCO2A1), a high-affinity carrier for prostaglandins (PGs), to the parturition process. At gestational day (GD) 15.5, OATP2A1 is co-localized with 15-hydroxy-PG dehydrogenase in the mouse placental junctional zone and facilitates PG degradation by delivering PGs to the cytoplasm. Slco2a1 (+/-) females mated with Slco2a1 (-/-) males frequently showed elevated circulating progesterone at GD18.5 and delayed parturition. Progesterone receptor inhibition by RU486 treatment at GD18.5 blocked the delay of parturition. In the junctional zone, PGE2 stimulated placental lactogen II (PL-II) production, resulting in higher expression of PL-II in Slco2a1 (-/-) placenta at GD18.5. Indomethacin treatment at GD15.5 suppressed the PL-II overproduction at GD18.5 in Slco2a1 (-/-) embryo-bearing dams, which promoted progesterone withdrawal and corrected the delayed parturition. These results suggest that extracellular PGE2 reduction by OATP2A1 at mid-pregnancy would be associated with progesterone withdrawal by suppressing PL-II production, triggering parturition onset.
Collapse
Affiliation(s)
- Mai Inagaki
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | | | - Takeo Nakanishi
- Faculty of Pharmacy, Takasaki University of Health and Welfare, Takasaki, Gunma 370-0033, Japan; Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Hiroaki Shimada
- Faculty of Pharmacy, Kindai University, Higashiosaka, Osaka 577-8502, Japan
| | - Saki Noguchi
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Shin-Ichi Akanuma
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Masanori Tachikawa
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Ken-Ichi Hosoya
- Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| | - Emi Nakashima
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo 105-8512, Japan
| | - Masatoshi Tomi
- Faculty of Pharmacy, Keio University, Minato-ku, Tokyo 105-8512, Japan.
| |
Collapse
|
50
|
Wahedi A, Günther A, Weyrich A, Sondheimer N. The mitochondrial genome of Cavia aperea. MITOCHONDRIAL DNA PART B-RESOURCES 2020; 5:2147-2148. [PMID: 33457761 PMCID: PMC7782685 DOI: 10.1080/23802359.2020.1768918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Cavia aperea is a wild guinea pig found throughout South America. The previously published mitochondrial sequence for C. aperea was highly divergent from the C. porcellus sequence and contained stop codons within open reading frames. Here we resequenced the mitochondrial genomes of C. aperea and C. porcellus. Both sequences reflect gene organization typical for mammalian mitochondrial DNA. Our C. aperea mtDNA sequence shows that all of the open reading frames are intact, but confirms the strikingly low level of sequence identity (92.7%) with the closely related C. porcellus mtDNA.
Collapse
Affiliation(s)
- Azizia Wahedi
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,The Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Anja Günther
- Max Planck Research Group Behavioural Ecology of Individual Differences, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Alexandra Weyrich
- Leibniz Institute for Zoo and Wildlife Research (IZW), Berlin, Germany
| | - Neal Sondheimer
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.,The Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|