1
|
Cho HY, Park KH, Oh E, Lee MJ, Choi BY, Im EM. Plasma acute phase proteins as potential predictors of intra-amniotic inflammation and infection in preterm premature rupture of membranes. Innate Immun 2024:17534259241306237. [PMID: 39711480 DOI: 10.1177/17534259241306237] [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: 12/24/2024] Open
Abstract
BACKGROUND We aimed to investigate the potential of altered levels of various acute phase proteins (APPs) in the plasma, either used alone or in combination with ultrasound-, clinical-, and conventional blood-based tests, for predicting the risk of intra-amniotic inflammation (IAI), microbial invasion of the amniotic cavity (MIAC), histologic chorioamnionitis (HCA), and funisitis in women with preterm premature rupture of membranes (PPROM). METHODS A total of 195 consecutive pregnancies involving singleton women with PPROM (at 23 + 0-34 + 0 weeks) who underwent amniocentesis and from whom plasma samples were obtained at amniocentesis were retrospectively included in this study. Amniotic fluid (AF) was cultured to assess the MIAC and analyzed for interleukin (IL)-6 levels to define IAI (AF IL-6 level of ≥2.6 ng/mL). The plasma concentrations of hepcidin, mannose-binding lectin (MBL), pentraxin-2, retinol-binding protein 4 (RBP4), serum amyloid A1 (SAA1), and serpin A1 were determined using ELISA. Ultrasonographic cervical length (CL), neutrophil-to-lymphocyte ratio (NLR), and C-reactive protein levels were measured. IAI/MIAC was defined as IAI, MIAC, or both. RESULTS Multivariate logistic regression analyses showed the following: (1) elevated plasma levels of hepcidin and SAA1 and decreased levels of RBP4 in the plasma were independently associated with IAI/MIAC and (2) decreased plasma RBP4 levels were independently associated with funisitis; however, (3) none of the plasma APPs investigated were associated with acute HCA when adjusted for baseline covariates. Using stepwise regression analysis, noninvasive prediction models comprising plasma RBP4 levels, CL, NLR, and gestational age at sampling were proposed, which provided a good prediction of IAI/MIAC and funisitis (area under the curve: 0.80 and 0.72, respectively). CONCLUSIONS Hepcidin, RBP4, and SAA1 were identified as potential APP biomarkers in the plasma predictive of IAI/MIAC or funisitis in patients with PPROM. In particular, combination of these APP biomarkers with ultrasound-, clinical-, and conventional blood-based markers can significantly support the diagnosis of IAI/MIAC and funisitis.
Collapse
Affiliation(s)
- Hee Young Cho
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
- Institute of Reproductive Medicine and Population, Medical Research Center, Seoul National University, Seoul, Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Eunji Oh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Bo Young Choi
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Eun Mi Im
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
2
|
Arslan Z. Microchimerism: The mystery of multiple DNA and its implications in forensic sciences. Forensic Sci Int 2024; 367:112345. [PMID: 39675234 DOI: 10.1016/j.forsciint.2024.112345] [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: 11/15/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 12/17/2024]
Abstract
Microchimerism (MC) refers to the presence of small amounts of foreign cells or DNA in the tissues or circulation of an individual. It generally occurs through mother-fetus interaction, twin pregnancies, and intergenerational transmission. MC is influenced by genetic and environmental factors such as toxic conditions, immunological suppression, and various diseases (influenza, COVID-19, etc.). Progenitor cells transferred from the fetus to the mother through fetal MC are known to differentiate into neurons in the maternal brain. Although the relationship between these cells and the brain is not fully understood, it is thought that they may play a role in the emergence of some mental illnesses. The long-term presence of microchimeric cells in the body by differentiating into various cell types such as the brain, heart, bone, liver, and lung can lead to the presence of two or more DNA sets in an individual. This can lead to confusion in forensic identification and sex determination processes. This review aims to provide a comprehensive review of the definition, transmission pathways, detection duration in the human body, associated diseases, analytical detection techniques, and the importance of MC in forensic sciences. In this context, it is aimed to draw attention to the potential dangers of MC and contribute to the justice system. Furthermore, this study emphasizes the need for scientific research on this topic by creating a starting point for future research in the field of MC.
Collapse
Affiliation(s)
- Zeynep Arslan
- Department of Science, Institute of Forensic Sciences and Legal Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey.
| |
Collapse
|
3
|
Kammala AK, Lintao RCV, Hoy R, Selim J, Luisi J, Yaklic JL, Ameredes BT, Menon R. Fetal microchimeric cells influence maternal lung health following term and preterm births. Sci Rep 2024; 14:28417. [PMID: 39557969 PMCID: PMC11574256 DOI: 10.1038/s41598-024-79795-0] [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: 08/06/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024] Open
Abstract
Fetal microchimerism, the presence of fetal cells in maternal tissues, has garnered interest for its potential role in maternal physiology. In this study, we aimed to explore the impact of fetal microchimeric cells on maternal lung health following term and preterm delivery, particularly in the context of infection-induced preterm birth and subsequent allergic challenges. We characterized the immune cells in maternal lungs using a transgenic mouse model (mT+ Ve, Td Tomato) and high dimensional mass cytometry (CyTOF) techniques. We evaluated their influence on lung function and inflammation. Our findings revealed distinct differences in the immune cell composition of maternal lungs between term and preterm deliveries. Mice delivered preterm significantly increased in fetal-specific cells, such as activated macrophages and Tbet + Ve memory B-cells, compared to term-delivered mice. Conversely, term deliveries showed elevated levels of CD4 cells. Furthermore, preterm-delivered dams demonstrated heightened airway hyperresponsiveness, pro-inflammatory cytokine expression, cellular infiltration, and lung mucous production compared to term-delivered dams. Co-culture experiments demonstrated that microchimeric cells from preterm births stimulated the production of inflammatory cytokines IL-6 and TNF-α in lung epithelial cells. These findings shed light on the complex immune dynamics postpartum and their role in lung complications after preterm birth. Understanding these mechanisms could provide insights for targeted interventions to improve maternal lung health in at-risk populations.
Collapse
Affiliation(s)
- Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| | - Ryan C V Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Richa Hoy
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jessica Selim
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA
- John Sealy School of Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jonathan Luisi
- Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jerome L Yaklic
- Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Bill T Ameredes
- Department of Internal Medicine, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX, 77555-1062, USA.
| |
Collapse
|
4
|
McCoy DE, Haig D, Kotler J. Egg donation and gestational surrogacy: Pregnancy is riskier with an unrelated embryo. Early Hum Dev 2024; 196:106072. [PMID: 39106717 DOI: 10.1016/j.earlhumdev.2024.106072] [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: 04/29/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 08/09/2024]
Abstract
Modern medicine has revolutionized family planning. Remarkably, women1 can carry to term embryos with whom they share no genetic connection, a feat made possible through egg donation and/or gestational surrogacy. Our reproductive systems evolved to accommodate embryos that are 50% related to the carrier, not 0% related. Here, we apply evolutionary theory to explain how and why pregnancy is riskier with an unrelated embryo. When a woman gestates an unrelated embryo, she is significantly more likely to develop preeclampsia and other diseases above and beyond the known risks associated with advanced maternal age, IVF, multiple gestation, and subfertility. Such "allogeneic pregnancies" are riskier even in fertile, healthy, commercial surrogates and when the egg is donated by a young, healthy donor. We propose that unrelated embryos present a special immune challenge to the gestational carrier, because they have fewer matching genes to the maternal body-therefore exacerbating symptoms of evolutionary maternal-fetal conflict. Indeed, maternal risks seem lower when the embryo is more related to the carrier, e.g., if a sister donates the egg. Finally, we discuss microchimerism in egg donation pregnancies, whereby wholly foreign cells pass from mother to embryo and vice-versa. We conclude with several medical proposals. First, egg donors and surrogates should be informed of the increased health risks they would face. In considerations of risk, these young, fertile women should not be compared to older, infertile women undergoing IVF; the proper comparison group is other young, fertile women. Second, contrary to some medical advice, perhaps genetically-related egg donors and surrogates should be preferred, all else equal. An immunological matching scheme, like what is used for organ transplants, could improve surrogate pregnancy outcomes. Third, more research is needed on microchimerism, sperm exposure, and the long-term impacts of allogeneic pregnancies on maternal and child health.
Collapse
Affiliation(s)
- Dakota E McCoy
- Department of Ecology and Evolution, The University of Chicago, Chicago, IL 60637, United States of America; Marine Biological Laboratory, Woods Hole, MA 02543, United States of America.
| | - David Haig
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02136, United States of America
| | - Jennifer Kotler
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02136, United States of America.
| |
Collapse
|
5
|
Park KH, Lee KN, Cho I, Lee MJ, Choi BY, Jeong DE. Plasma Kallistatin and Progranulin as Predictive Biomarkers of Intraamniotic Inflammation, Microbial Invasion of the Amniotic Cavity, and Composite Neonatal Morbidity/Mortality in Women With Preterm Premature Rupture of Membranes. Am J Reprod Immunol 2024; 92:e13909. [PMID: 39072836 DOI: 10.1111/aji.13909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/03/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
PROBLEM To explore the clinical utility of nine inflammatory immune-, adhesion-, and extracellular matrix-related mediators in the plasma for predicting intraamniotic inflammation and/or microbial invasion of the amniotic cavity (IAI/MIAC) and composite neonatal morbidity and/or mortality (CNMM) in women with preterm premature rupture of membranes (PPROM) when used alone or in combination with conventional blood-, ultrasound-, and clinical-based factors. METHODS OF STUDY This retrospective cohort comprised 173 singleton pregnant women with PPROM (24 + 0 - 33 + 6 weeks), who underwent amniocentesis. Amniotic fluid was cultured for microorganisms and assayed for IL-6 levels. Plasma levels of AFP, CXCL14, E-selectin, Gal-3BP, kallistatin, progranulin, P-selectin, TGFBI, and VDBP were determined by ELISA. Ultrasonographic cervical length (CL) and neutrophil-to-lymphocyte ratio (NLR) were measured. RESULTS Multivariate logistic regression analyses revealed significant associations between (i) decreased plasma kallistatin levels and IAI/MIAC and (ii) decreased plasma progranulin levels and increased CNMM risk after adjusting for baseline variables (e.g., gestational age at sampling [or delivery] and parity). Using stepwise regression analysis, noninvasive prediction models for IAI/MIAC and CNMM risks were developed, which included plasma progranulin levels, NLR, CL, and gestational age at sampling, and provided a good prediction of the corresponding endpoints (area under the curve: 0.79 and 0.87, respectively). CONCLUSIONS Kallistatin and progranulin are potentially valuable plasma biomarkers for predicting IAI/MIAC and CNMM in women with PPROM. Particularly, the combination of these plasma biomarkers with conventional blood-, ultrasound-, and clinical-based factors can significantly support the diagnosis of IAI/MIAC and CNMM.
Collapse
Affiliation(s)
- Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Chungnam National University Hospital, Daejeon, South Korea
| | - Iseop Cho
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Bo Young Choi
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Da Eun Jeong
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| |
Collapse
|
6
|
Joo E, Hong S, Park KH, Kim HJ, Lee MJ, Shin S. Predictive potential of various plasma inflammation-, angiogenesis-, and extracellular matrix remodeling-associated mediators for intra-amniotic inflammation and/or microbial invasion of the amniotic cavity in preterm labor. Arch Gynecol Obstet 2024; 310:413-426. [PMID: 38329550 DOI: 10.1007/s00404-024-07378-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
PURPOSE To determine whether various inflammatory-, angiogenic/anti-angiogenic-, and extracellular matrix remodeling-associated proteins in plasma, alone or in combination with conventional blood-based markers, can predict intra-amniotic inflammation and/or microbial invasion of the amniotic cavity (IAI/MIAC) in women with spontaneous preterm labor (PTL). METHODS A total of 193 singleton pregnant women with PTL (23-33 weeks) were included in this retrospective cohort study. Plasma samples were obtained at the time of amniocentesis. Amniotic fluid (AF) was cultured for microorganism detection and consequent MIAC diagnosis. IL-6 levels were determined in AF and used to identify IAI (AF IL-6 ≥ 2.6 ng/mL). Endostatin, haptoglobin, IGFBP-2/3, LBP, M-CSF, MMP-2/8, pentraxin 3, PlGF, S100A8/A9, and VEGFR-1 levels were assayed in plasma samples by ELISA. CRP levels and neutrophil-to-lymphocyte ratio (NLR) were measured. RESULTS Plasma LBP, MMP-8, and S100A8/A9 levels, CRP levels, and NLR were significantly higher, and plasma IGFBP-2 and MMP-2 levels were significantly lower in women with IAI/MIAC than in those without this condition, whereas no baseline variables differed significantly between the two groups. Using a stepwise regression analysis, a noninvasive prediction model for IAI/MIAC was developed, which included plasma LBP, MMP-2, and MMP-8 levels (area under the curve [AUC], 0.785). The AUC for this prediction model was significantly or borderline greater than that of any single factor included in the model. CONCLUSIONS IGFBP-2, LBP, MMP-2, MMP-8, and S100A8/A9 may represent valuable plasma biomarkers for predicting IAI/MIAC in women with PTL. Combination of LBP, MMP-2, and MMP-8 expression data can significantly improve the predictive potential for IAI/MIAC.
Collapse
Affiliation(s)
- Eunwook Joo
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Seongnam, Kyeonggido, 463-707, Korea
| | - Subeen Hong
- Department of Obstetrics and Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Seongnam, Kyeonggido, 463-707, Korea.
| | - Hyeon Ji Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Seongnam, Kyeonggido, 463-707, Korea
| | - Min Jung Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173 Beon-gil, Seongnam, Kyeonggido, 463-707, Korea
| | - Sue Shin
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul National University Boramae Hospital, Seoul, Korea
| |
Collapse
|
7
|
Fitzpatrick G, Huang Y, Qiu F, Habgood MD, Medcalf RL, Ho H, Dziegielewska KM, Saunders NR. Entry of cannabidiol into the fetal, postnatal and adult rat brain. Cell Tissue Res 2024; 396:177-195. [PMID: 38366086 PMCID: PMC11055756 DOI: 10.1007/s00441-024-03867-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/22/2024] [Indexed: 02/18/2024]
Abstract
Cannabidiol is a major component of cannabis but without known psychoactive properties. A wide range of properties have been attributed to it, such as anti-inflammatory, analgesic, anti-cancer, anti-seizure and anxiolytic. However, being a fairly new compound in its purified form, little is known about cannabidiol brain entry, especially during development. Sprague Dawley rats at four developmental ages: embryonic day E19, postnatal day P4 and P12 and non-pregnant adult females were administered intraperitoneal cannabidiol at 10 mg/kg with [3H] labelled cannabidiol. To investigate the extent of placental transfer, the drug was injected intravenously into E19 pregnant dams. Levels of [3H]-cannabidiol in blood plasma, cerebrospinal fluid and brain were estimated by liquid scintillation counting. Plasma protein binding of cannabidiol was identified by polyacrylamide gel electrophoresis and its bound and unbound fractions measured by ultrafiltration. Using available RNA-sequencing datasets of E19 rat brain, choroid plexus and placenta, as well as P5 and adult brain and choroid plexus, expression of 13 main cannabidiol receptors was analysed. Results showed that cannabidiol rapidly entered both the developing and adult brains. Entry into CSF was more limited. Its transfer across the placenta was substantially restricted as only about 50% of maternal blood plasma cannabidiol concentration was detected in fetal plasma. Albumin was the main, but not exclusive, cannabidiol binding protein at all ages. Several transcripts for cannabidiol receptors were expressed in age- and tissue-specific manner indicating that cannabidiol may have different functional effects in the fetal compared to adult brain.
Collapse
Affiliation(s)
- Georgia Fitzpatrick
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Yifan Huang
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Fiona Qiu
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Mark D Habgood
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Robert L Medcalf
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - Heidi Ho
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | | | - Norman R Saunders
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
8
|
Fjeldstad HE, Jacobsen DP, Johnsen GM, Sugulle M, Chae A, Kanaan SB, Gammill HS, Staff AC. Fetal-origin cells in maternal circulation correlate with placental dysfunction, fetal sex, and severe hypertension during pregnancy. J Reprod Immunol 2024; 162:104206. [PMID: 38309014 DOI: 10.1016/j.jri.2024.104206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/21/2023] [Accepted: 01/17/2024] [Indexed: 02/05/2024]
Abstract
Fetal microchimerism (FMc) arises when fetal cells enter maternal circulation, potentially persisting for decades. Increased FMc is associated with fetal growth restriction, preeclampsia, and anti-angiogenic shift in placenta-associated proteins in diabetic and normotensive term pregnancies. The two-stage model of preeclampsia postulates that placental dysfunction causes such shift in placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFLt-1), triggering maternal vascular inflammation and endothelial dysfunction. We investigated whether anti-angiogenic shift, fetal sex, fetal growth restriction, and severe maternal hypertension correlate with FMc in hypertensive disorders of pregnancy with new-onset features (n = 125). Maternal blood was drawn pre-delivery at > 25 weeks' gestation. FMc was detected by quantitative polymerase chain reaction targeting paternally inherited unique fetal alleles. PlGF and sFlt-1 were measured by immunoassay. We estimated odds ratios (ORs) by logistic regression and detection rate ratios (DRRs) by negative binomial regression. PlGF correlated negatively with FMc quantity (DRR = 0.2, p = 0.005) and female fetal sex correlated positively with FMc prevalence (OR = 5.0, p < 0.001) and quantity (DRR = 4.5, p < 0.001). Fetal growth restriction no longer correlated with increased FMc quantity after adjustment for correlates of placental dysfunction (DRR = 1.5, p = 0.272), whereas severe hypertension remained correlated with both FMc measures (OR = 5.5, p = 0.006; DRR = 6.3, p = 0.001). Our findings suggest that increased FMc is independently associated with both stages of the two-stage preeclampsia model. The association with female fetal sex has implications for microchimerism detection methodology. Future studies should target both male and female-origin FMc and focus on clarifying which placental mechanisms impact fetal cell transfer and how FMc impacts the maternal vasculature.
Collapse
Affiliation(s)
- Heidi E Fjeldstad
- Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway.
| | - Daniel P Jacobsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Guro M Johnsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| | - Angel Chae
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Obstetrics and Gynecology Research Division, University of Washington, Seattle, WA, USA
| | - Sami B Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Chimerocyte, Inc., Seattle, WA, USA
| | - Hilary S Gammill
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Obstetrics and Gynecology Research Division, University of Washington, Seattle, WA, USA
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Burton GJ. Fetal microchimerism, pregnancy epiphenomenon or kinship indicator? Proc Biol Sci 2023; 290:20231906. [PMID: 37817590 PMCID: PMC10565412 DOI: 10.1098/rspb.2023.1906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Affiliation(s)
- Graham J. Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| |
Collapse
|
10
|
Yüzen D, Urbschat C, Schepanski S, Thiele K, Arck PC, Mittrücker H. Pregnancy-induced transfer of pathogen-specific T cells from mother to fetus in mice. EMBO Rep 2023; 24:e56829. [PMID: 37610043 PMCID: PMC10561172 DOI: 10.15252/embr.202356829] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/24/2023] Open
Abstract
Neonatal health is determined by the transfer of maternal antibodies from the mother to the fetus. Besides antibodies, maternal cells cross the placental barrier and seed into fetal organs. Contrary to maternal antibodies, maternal microchimeric cells (MMc) show a high longevity, as they can persist in the offspring until adulthood. Recent evidence highlights that MMc leukocytes promote neonatal immunity against early-life infections in mice and humans. As shown in mice, this promotion of immunity was attributable to an improved fetal immune development. Besides this indirect effect, MMc may be pathogen-specific and thus, directly clear pathogen threats in the offspring postnatally. By using ovalbumin recombinant Listeria monocytogenes (LmOVA), we here provide evidence that OVA-specific T cells are transferred from the mother to the fetus, which is associated with increased activation of T cells and a milder course of postnatal infection in the offspring. Our data highlight that maternally-derived passive immunity of the neonate is not limited to antibodies, as MMc have the potential to transfer immune memory between generations.
Collapse
Affiliation(s)
- Dennis Yüzen
- Division of Experimental Feto‐Maternal Medicine, Department of Obstetrics and Fetal MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
- Institute of ImmunologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christopher Urbschat
- Division of Experimental Feto‐Maternal Medicine, Department of Obstetrics and Fetal MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Steven Schepanski
- Division of Experimental Feto‐Maternal Medicine, Department of Obstetrics and Fetal MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Kristin Thiele
- Division of Experimental Feto‐Maternal Medicine, Department of Obstetrics and Fetal MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Petra C Arck
- Division of Experimental Feto‐Maternal Medicine, Department of Obstetrics and Fetal MedicineUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | | |
Collapse
|
11
|
Jacobsen DP, Fjeldstad HE, Sugulle M, Johnsen GM, Olsen MB, Kanaan SB, Staff AC. Fetal microchimerism and the two-stage model of preeclampsia. J Reprod Immunol 2023; 159:104124. [PMID: 37541161 DOI: 10.1016/j.jri.2023.104124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
Fetal cells cross the placenta during pregnancy and some have the ability to persist in maternal organs and circulation long-term, a phenomenon termed fetal microchimerism. These cells often belong to stem cell or immune cell lineages. The long-term effects of fetal microchimerism are likely mixed, potentially depending on the amount of fetal cells transferred, fetal-maternal histocompatibility and fetal cell-specific properties. Both human and animal data indicate that fetal-origin cells partake in tissue repair and may benefit maternal health overall. On the other hand, these cells have been implicated in inflammatory diseases by studies showing increased fetal microchimerism in women with autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. During pregnancy, preeclampsia is associated with increased cell-transfer between the mother and fetus, and an increase in immune cell subsets. In the current review, we discuss potential mechanisms of transplacental transfer, including passive leakage across the compromised diffusion barrier and active recruitment of cells residing in the placenta or fetal circulation. Within the conceptual framework of the two-stage model of preeclampsia, where syncytiotrophoblast stress is a common pathophysiological pathway to maternal and fetal clinical features of preeclampsia, we argue that microchimerism may represent a mechanistic link between stage 1 placental dysfunction and stage 2 maternal cardiovascular inflammation and endothelial dysfunction. Finally, we postulate that fetal microchimerism may contribute to the known association between placental syndromes and increased long-term maternal cardiovascular disease risk. Fetal microchimerism research represents an exciting opportunity for developing new disease biomarkers and targeted prophylaxis against maternal diseases.
Collapse
Affiliation(s)
- Daniel P Jacobsen
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway.
| | | | - Meryam Sugulle
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Guro M Johnsen
- Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Maria B Olsen
- Faculty of Medicine, University of Oslo, Oslo, Norway; Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Sami B Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Chimerocyte, Inc., Seattle, WA, USA
| | - Anne Cathrine Staff
- Faculty of Medicine, University of Oslo, Oslo, Norway; Division of Obstetrics and Gynaecology, Oslo University Hospital, Ullevål, Oslo, Norway
| |
Collapse
|
12
|
Úbeda F, Wild G. Microchimerism as a source of information on future pregnancies. Proc Biol Sci 2023; 290:20231142. [PMID: 37608718 PMCID: PMC10445024 DOI: 10.1098/rspb.2023.1142] [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: 05/23/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023] Open
Abstract
Small numbers of fetal cells cross the placenta during pregnancy turning mothers into microchimeras. Fetal cells from all previous pregnancies accumulate forming the mother's fetal microchiome. What is significant about microchimeric cells is that they have been linked to health problems including reproductive and autoimmune diseases. Three decades after the discovery of fetal microchimerism, the function of these cells remains a mystery. Here, we contend that the role of microchimeric cells is to inform the fetus about the likelihood that its genes are present in future pregnancies. We argue that, when genes are more likely than average to be in future maternal siblings, fetuses will send a fixed number of cells that will not elicit a maternal immune response against them. However, when genes are less likely to be in future maternal siblings, fetuses will send an ever-increasing number of cells that will elicit an ever-stronger maternal immune response. Our work can explain the observed clinical association between microchimeric cells and pre-eclampsia. However, our work predicts that this association should be stronger in women with a genetically diverse microchiome. If supported by medical tests, our work would allow establishing the likelihood of pregnancy or autoimmune problems advising medical interventions.
Collapse
Affiliation(s)
- Francisco Úbeda
- Department of Biological Sciences, Royal Holloway, University of London, Egham TW20 0EX, UK
| | - Geoff Wild
- Department of Mathematics, The University of Western Ontario, London, Ontario, Canada N6A 5B7
| |
Collapse
|
13
|
Bahrami H, Tafrihi M. Global trends of cancer: The role of diet, lifestyle, and environmental factors. CANCER INNOVATION 2023; 2:290-301. [PMID: 38089751 PMCID: PMC10686168 DOI: 10.1002/cai2.76] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/13/2023] [Accepted: 04/18/2023] [Indexed: 10/15/2024]
Abstract
Effective treatment of cancer requires understanding the nature of the disease and accurately addressing the main root causes. General risk factors for cancer include poor nutrition, an acidogenic diet, an unhealthy lifestyle, and exposure to carcinogens such as toxins, chemicals, and radiation. The risk of developing cancers may be reduced by sufficient oxygenation and maintaining optimal alkalinity and nutritional balance at the cell level. The review paper summarizes some diet and lifestyle modifications that may potentially be considered for preventing and controlling some cancers. Moreover, worldwide statistical data for cancer incidence rates published by International Agency for Research on Cancer are analyzed for certain cancers regionally, concerning the effect of dietary habits and environmental factors that meaningfully correlate with the global trends of cancer. The study of cancer root causes integrated with analyzing the statistics related to cancer incidence rates suggests that the risk of developing cancer may be reduced by modifying dietary habits and lifestyle factors, as well as reducing exposure to carcinogens. Those with healthy balanced dietary habits may have a lower cancer risk than those who frequently have unhealthy diets; hence, considering a balanced natural diet and healthy lifestyle may be suggested as a complementary or alternative solution in cancer treatments.
Collapse
Affiliation(s)
| | - Majid Tafrihi
- Molecular and Cell Biology Research Laboratory 2, Department of Molecular and Cell Biology, Faculty of SciencesUniversity of MazandaranBabolsarIran
| |
Collapse
|
14
|
Wolfova K, Wu D, Weiss J, Cermakova P, Kohler HP, Skirbekk VF, Stern Y, Gemmill A, Tom SE. Sons and parental cognition in mid-life and older adulthood. J Psychiatr Res 2022; 156:284-290. [PMID: 36279678 PMCID: PMC10103684 DOI: 10.1016/j.jpsychires.2022.10.026] [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: 07/05/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Prior research suggests a relationship between number of sons and maternal long-term health outcomes, including dementia. We assessed the relationship between having sons and parental cognitive aging. Specifically, we investigated the relationship between having at least 1 son and parental baseline cognition level and rate of cognitive decline, accounting for life course sociodemographic characteristics in a cohort of 13 222 adults aged ≥50 years from the US Health and Retirement Study. We included only participants with at least one child. We further explored whether this relationship varies by parental sex and whether the magnitude of the relationship increases with each additional son. Cognition was assessed biennially for a maximum of nine times as a sum of scores from immediate and delayed 10-noun free recall tests, a serial 7s subtraction test, and a backwards counting test. Associations were evaluated using linear mixed-effects models, stepwise adjusting for sociodemographic and health-related factors. In our analytic sample of parents, a total of 82.3% of respondents had at least 1 son and 61.6% of respondents were female. Parents of at least 1 son had a faster rate of cognitive decline in comparison to parents without any son. Our results also suggest that cognitive decline was faster among parents of multiple sons, compared to parents with only daughters. Thus, the results support the theory that having sons might have a long-term negative effect on parental cognition.
Collapse
Affiliation(s)
- Katrin Wolfova
- Department of Psychiatry and Medical Psychology, Third Faculty of Medicine, Charles University, Prague, 100 00, Czech Republic; National Institute of Mental Health, Klecany, 250 67, Czech Republic; Department of Neurology, Columbia University, 10032, New York, USA
| | - Di Wu
- Department of Biostatistics, Columbia University, 10032, New York, USA
| | - Jordan Weiss
- Department of Demography, UC Berkeley, Berkeley, CA, 94720, USA
| | - Pavla Cermakova
- National Institute of Mental Health, Klecany, 250 67, Czech Republic; Department of Epidemiology, Second Faculty of Medicine, Charles University, Prague, 150 06, Czech Republic
| | - Hans-Peter Kohler
- Department of Sociology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vegard Fykse Skirbekk
- Centre for Fertility and Health, The Norwegian Institute of Public Health, Oslo, 0473, Norway
| | - Yaakov Stern
- Department of Neurology, Columbia University, 10032, New York, USA
| | - Alison Gemmill
- Department of Population, Family and Reproductive Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, 21205, Maryland, USA
| | - Sarah E Tom
- Department of Neurology, Columbia University, 10032, New York, USA; Department of Epidemiology, Columbia University, 10032, New York, USA.
| |
Collapse
|
15
|
Lee JE, Dan K, Kim HJ, Kim YM, Park KH. Plasma proteomic analysis to identify potential biomarkers of histologic chorioamnionitis in women with preterm premature rupture of membranes. PLoS One 2022; 17:e0270884. [PMID: 35797368 PMCID: PMC9262229 DOI: 10.1371/journal.pone.0270884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 06/18/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction
To identify potential biomarkers in the plasma that could predict histologic chorioamnionitis (HCA) in women with preterm premature rupture of membranes (PPROM), using shotgun and targeted proteomic analyses.
Methods
This retrospective cohort study included 78 singleton pregnant women with PPROM (24–34 gestational weeks) who delivered within 96 h of blood sampling. Maternal plasma samples were analyzed by label-free liquid chromatography-tandem mass spectrometry for proteome profiling in a nested case-control study design (HCA cases vs. non-HCA controls [n = 9 each]). Differential expression of 12 candidate proteins was assessed by multiple reaction monitoring-mass spectrometry (MRM-MS) analysis in individual plasma samples from cases and controls matched by gestational age at sampling (n = 40, cohort 1). A validation study was further performed in an independent study group (n = 38, cohort 2) using ELISA and turbidimetric immunoassay for three differentially expressed proteins.
Results
Shotgun proteomics analyses yielded 18 proteins that were differentially expressed (P < 0.05) between HCA cases and non-HCA controls. MRM-MS analysis of 12 differentially expressed proteins further revealed that the CRP, C4A, and SAA4 levels were significantly increased in women with HCA. A multi-marker panel comprising plasma SAA4 and C4A showed enhanced potential for differentiating HCA from non-HCA women (area under the curve = 0.899). Additional validation of these findings by ELISA assays revealed that the CRP levels were significantly higher in women with HCA than in those without HCA, whereas the plasma levels of C4A and SAA4 did not significantly differ between the two groups.
Conclusions
Plasma C4A, SAA4, and CRP were identified as potential biomarkers for detecting HCA in women with PPROM, based on targeted and shotgun proteomic analyses, showing good accuracy when used as a combined dual-biomarker panel (C4A and SAA4). Nevertheless, ELISA validation of these proteins, except for CRP, may not yield clinically useful markers for predicting HCA.
Collapse
Affiliation(s)
- Ji Eun Lee
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Korea
| | - Kisoon Dan
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyeon Ji Kim
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Mi Kim
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyo Hoon Park
- Departments of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail:
| |
Collapse
|
16
|
Romanis EC. Assisted gestative technologies. JOURNAL OF MEDICAL ETHICS 2022; 48:439-446. [PMID: 35277466 DOI: 10.1136/medethics-2021-107769] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 02/17/2022] [Indexed: 06/14/2023]
Abstract
A large body of literature considers the ethico-legal and regulatory issues surrounding assisted conception. Surrogacy, however, within this body of literature is an odd-fit. It involves a unique demand of another person-a form of reproductive labour-that many other aspects of assisted conception, such as gamete donation do not involve. Surrogacy is a form of assisted gestation. The potential alternatives for individuals who want a genetically related child but who do not have the capacity to gestate are ever increasing: with the advent of uterus transplantation (UTx), and the anticipated development of artificial placentas.In this paper, I highlight the pertinent conceptual differences between technologies assisting conception and those assisting gestation to demonstrate that distinct issues arise when assisted gestative technologies (AGTs) are used. I argue that there is utility in considering AGTs as a genus of technologies. These technologies perform the same function from the perspective of putative parents and might exist on a spectrum of alternatives when they become more available. Moreover, since many of the same or very similar ethical, social and legal issues are raised by surrogacy, UTx and ectogestation, analysis of these issues will be better contextualised by considering these technologies together. Many of the matters currently being highlighted for reform in the context of surrogacy will inevitably impact on how other forms of assisted gestation are governed. The conversation should be broadened; we should consider how far other emerging technologies might be accommodated in revised regulatory schemes.
Collapse
|
17
|
Murrieta-Coxca JM, Fuentes-Zacarias P, Ospina-Prieto S, Markert UR, Morales-Prieto DM. Synergies of Extracellular Vesicles and Microchimerism in Promoting Immunotolerance During Pregnancy. Front Immunol 2022; 13:837281. [PMID: 35844513 PMCID: PMC9285877 DOI: 10.3389/fimmu.2022.837281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of biological identity has been traditionally a central issue in immunology. The assumption that entities foreign to a specific organism should be rejected by its immune system, while self-entities do not trigger an immune response is challenged by the expanded immunotolerance observed in pregnancy. To explain this "immunological paradox", as it was first called by Sir Peter Medawar, several mechanisms have been described in the last decades. Among them, the intentional transfer and retention of small amounts of cells between a mother and her child have gained back attention. These microchimeric cells contribute to expanding allotolerance in both organisms and enhancing genetic fitness, but they could also provoke aberrant alloimmune activation. Understanding the mechanisms used by microchimeric cells to exert their function in pregnancy has proven to be challenging as per definition they are extremely rare. Profiting from studies in the field of transplantation and cancer research, a synergistic effect of microchimerism and cellular communication based on the secretion of extracellular vesicles (EVs) has begun to be unveiled. EVs are already known to play a pivotal role in feto-maternal tolerance by transferring cargo from fetal to maternal immune cells to reshape their function. A further aspect of EVs is their function in antigen presentation either directly or on the surface of recipient cells. Here, we review the current understanding of microchimerism in the feto-maternal tolerance during human pregnancy and the potential role of EVs in mediating the allorecognition and tropism of microchimeric cells.
Collapse
Affiliation(s)
| | | | | | - Udo R. Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | | |
Collapse
|
18
|
Hustvedt S. Umbilical phantoms. THE INTERNATIONAL JOURNAL OF PSYCHOANALYSIS 2022; 103:368-380. [PMID: 35440273 DOI: 10.1080/00207578.2021.2014118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
19
|
Meiotic drive in chronic lymphocytic leukemia compared with other malignant blood disorders. Sci Rep 2022; 12:6138. [PMID: 35413962 PMCID: PMC9005523 DOI: 10.1038/s41598-022-09602-1] [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: 11/23/2021] [Accepted: 03/22/2022] [Indexed: 11/12/2022] Open
Abstract
The heredity of the malignant blood disorders, leukemias, lymphomas and myeloma, has so far been largely unknown. The present study comprises genealogical investigations of one hundred and twelve Scandinavian families with unrelated parents and two or more cases of malignant blood disease. For comparison, one large family with related family members and three hundred and forty-one cases of malignant blood disease from the Faroese population was included. The inheritance is non-Mendelian, a combination of genomic parental imprinting and feto-maternal microchimerism. There is significantly more segregation in maternal than in paternal lines, predominance of mother-daughter combinations in maternal lines, and father-son combinations in paternal lines. Chronic lymphocytic leukemia is the most frequent diagnosis in the family material, and chronic lymphocytic leukemia has a transgenerational segregation that is unique in that inheritance of susceptibility to chronic lymphocytic leukemia is predominant in males of paternal lines. Male offspring with chronic lymphocytic leukemia in paternal lines have a birth-order effect, which is manifest by the fact that there are significantly more male patients late in the sibling line. In addition, there is contravariation in chronic lymphocytic leukemia, i.e. lower occurrence than expected in relation to other diagnoses, interpreted in such a way that chronic lymphocytic leukemia remains isolated in the pedigree in relation to other diagnoses of malignant blood disease. Another non-Mendelian function appears in the form of anticipation, i.e. increased intensity of malignancy down through the generations and a lower age at onset of disease than otherwise seen in cases from the Cancer Registers, in acute lymphoblastic leukemia, for example. It is discussed that this non-Mendelian segregation seems to spread the susceptibility genes depending on the gender of the parents and not equally to all children in the sibling line, with some remaining unaffected by susceptibility i.e. "healthy and unaffected", due to a birth order effect. In addition, anticipation is regarded as a non-Mendelian mechanism that can amplify, «preserve» these vital susceptibility genes in the family. Perhaps this segregation also results in a sorting of the susceptibility, as the percentage of follicular lymphoma and diffuse large B-cell lymphoma is lower in the family material than in an unselected material. Although leukemias, lymphomas and myelomas are potentially fatal diseases, this non-Mendelian distribution and amplification hardly play any quantitative role in the survival of Homo sapiens, because these diseases mostly occur after fertile age.
Collapse
|
20
|
|
21
|
Cómitre-Mariano B, Martínez-García M, García-Gálvez B, Paternina-Die M, Desco M, Carmona S, Gómez-Gaviro MV. Feto-maternal microchimerism: Memories from pregnancy. iScience 2022; 25:103664. [PMID: 35072002 PMCID: PMC8762399 DOI: 10.1016/j.isci.2021.103664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is a bidirectional transplacental cell trafficking between mother and fetus during pregnancy in placental mammals. The presence and persistence of fetal cells in maternal tissues are known as fetal microchimerism (FMc). FMc has high multilineage potential with a great ability to differentiate and functionally integrate into maternal tissue. FMc has been found in various maternal tissues in animal models and humans. Its permanence in the maternal body up to decades after delivery suggests it might play an essential role in maternal pathophysiology. Studying the presence, localization, and characteristics of FMc in maternal tissues is key to understanding its impact on the woman's body. Here we comprehensively review the existence of FMc in different species and organs and tissues, aiming to better characterize their possible role in human health and disease. We also highlight several methodological considerations that would optimize the detection, quantification, and functional determination of FMc.
Collapse
Affiliation(s)
- Blanca Cómitre-Mariano
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - Magdalena Martínez-García
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), C/ Monforte de Lemos 3-5, Instituto de Salud Carlos III, Pabellón 11, planta baja, 28029 Madrid, Spain
| | - Bárbara García-Gálvez
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain
| | - María Paternina-Die
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), C/ Monforte de Lemos 3-5, Instituto de Salud Carlos III, Pabellón 11, planta baja, 28029 Madrid, Spain
| | - Manuel Desco
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), C/ Monforte de Lemos 3-5, Instituto de Salud Carlos III, Pabellón 11, planta baja, 28029 Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Avenida de la Universidad, 30, 28911 Leganés, Spain.,Centro Nacional de Investigaciones Cardiovasculares Carlos III, CNIC, C/ Melchor Fernandez Almagro 3, 28029 Madrid, Spain
| | - Susanna Carmona
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), C/ Monforte de Lemos 3-5, Instituto de Salud Carlos III, Pabellón 11, planta baja, 28029 Madrid, Spain
| | - María Victoria Gómez-Gaviro
- Instituto de Investigación Sanitaria Gregorio Marañón. (IiSGM), C/Doctor Esquerdo 46, 28007 Madrid, Spain.,Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), C/ Monforte de Lemos 3-5, Instituto de Salud Carlos III, Pabellón 11, planta baja, 28029 Madrid, Spain.,Departamento de Bioingeniería e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, Avenida de la Universidad, 30, 28911 Leganés, Spain
| |
Collapse
|
22
|
Chiou SYS, Kysenius K, Huang Y, Habgood MD, Koehn LM, Qiu F, Crouch PJ, Varshney S, Ganio K, Dziegielewska KM, Saunders NR. Lithium administered to pregnant, lactating and neonatal rats: entry into developing brain. Fluids Barriers CNS 2021; 18:57. [PMID: 34876168 PMCID: PMC8650431 DOI: 10.1186/s12987-021-00285-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 11/02/2021] [Indexed: 12/04/2022] Open
Abstract
Background Little is known about the extent of drug entry into developing brain, when administered to pregnant and lactating women. Lithium is commonly prescribed for bipolar disorder. Here we studied transfer of lithium given to dams, into blood, brain and cerebrospinal fluid (CSF) in embryonic and postnatal animals as well as adults. Methods Lithium chloride in a clinically relevant dose (3.2 mg/kg body weight) was injected intraperitoneally into pregnant (E15–18) and lactating dams (birth-P16/17) or directly into postnatal pups (P0–P16/17). Acute treatment involved a single injection; long-term treatment involved twice daily injections for the duration of the experiment. Following terminal anaesthesia blood plasma, CSF and brains were collected. Lithium levels and brain distribution were measured using Laser Ablation Inductively Coupled Plasma-Mass Spectrometry and total lithium levels were confirmed by Inductively Coupled Plasma-Mass Spectrometry. Results Lithium was detected in blood, CSF and brain of all fetal and postnatal pups following lithium treatment of dams. Its concentration in pups’ blood was consistently below that in maternal blood (30–35%) indicating significant protection by the placenta and breast tissue. However, much of the lithium that reached the fetus entered its brain. Levels of lithium in plasma fluctuated in different treatment groups but its concentration in CSF was stable at all ages, in agreement with known stable levels of endogenous ions in CSF. There was no significant increase of lithium transfer into CSF following application of Na+/K+ ATPase inhibitor (digoxin) in vivo, indicating that lithium transfer across choroid plexus epithelium is not likely to be via the Na+/K+ ATPase mechanism, at least early in development. Comparison with passive permeability markers suggested that in acute experiments lithium permeability was less than expected for diffusion but similar in long-term experiments at P2. Conclusions Information obtained on the distribution of lithium in developing brain provides a basis for studying possible deleterious effects on brain development and behaviour in offspring of mothers undergoing lithium therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00285-w.
Collapse
Affiliation(s)
- Shene Yi-Shiuan Chiou
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kai Kysenius
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yifan Huang
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Mark David Habgood
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Liam M Koehn
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Fiona Qiu
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Peter J Crouch
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Swati Varshney
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Katherine Ganio
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Katarzyna Magdalena Dziegielewska
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Norman Ruthven Saunders
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Neuroscience, Monash University, 99 Commercial Road, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
23
|
Fetomaternal hemorrhage: evaluation of recurrence within a large integrated healthcare system. Am J Obstet Gynecol 2021; 225:540.e1-540.e8. [PMID: 33961809 DOI: 10.1016/j.ajog.2021.04.257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND Fetomaternal hemorrhage is associated with severe fetal morbidity and mortality. The recurrence risk of fetomaternal hemorrhage is unknown. OBJECTIVE We sought to establish the recurrence rate of fetomaternal hemorrhage in a large integrated healthcare system over a 10-year period. STUDY DESIGN In this retrospective study within the Kaiser Permanente Northern California medical system, cases of fetomaternal hemorrhage were defined by either an elevated fetal hemoglobin level as determined by flow cytometry for a concerning pregnancy outcome (preterm delivery, perinatal demise, neonatal anemia, or transfusion within the first 2 days of life) or by perinatal demise with autopsy findings suggestive of fetomaternal hemorrhage. The outcomes of subsequent pregnancies were reviewed for features of recurrence. RESULTS Within the 2008 to 2018 birth cohort of 375,864 pregnancies, flow cytometry testing for fetal hemoglobin levels was performed in 20,582 pregnancies. We identified 340 cases of fetomaternal hemorrhage (approximately 1 in 1100 births). Within the cohort of 340 affected pregnancies, perinatal loss was recorded for 80 (23.5%) pregnancies and 50 (14.7%) pregnancies delivered neonates who required transfusion. The affected patients had 225 subsequent pregnancies of which 210 were included in the analysis. Of these, 174 (82.9%) advanced beyond the threshold of viability and were delivered within our healthcare system. There was 1 case of recurrent fetomaternal hemorrhage identified. The recurrent case involved a spontaneous preterm delivery of an infant who was noted to have an elevated reticulocyte count but was clinically well. CONCLUSION Within our large integrated healthcare system, approximately 1 in 1100 pregnancies was affected by fetomaternal hemorrhage within a 10-year period, which is comparable with previous studies. We identified 1 case of recurrence, yielding a recurrence rate of 0.5%. This infant did not have features of clinically important fetomaternal hemorrhage. This information can inform counseling of patients with affected pregnancies.
Collapse
|
24
|
Murrieta-Coxca JM, Aengenheister L, Schmidt A, Markert UR, Buerki-Thurnherr T, Morales-Prieto DM. Addressing microchimerism in pregnancy by ex vivo human placenta perfusion. Placenta 2021; 117:78-86. [PMID: 34773744 DOI: 10.1016/j.placenta.2021.10.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 10/07/2021] [Accepted: 10/10/2021] [Indexed: 12/20/2022]
Abstract
The physical connection of mother and offspring during pregnancy allows the bi-directional exchange of a small number of cells through the placenta. These cells, which can persist long-term in the recipient individual are genetically foreign to it and therefore fulfill the principle of microchimerism. Over the last years, pioneer research on microchimeric cells revealed their role in immune adaptation during pregnancy and priming of tolerogenic responses in the progeny. However, the mechanisms involved in cell transfer across the placenta barrier remain poorly investigated. In this review, we summarize the evidence of fetomaternal microchimerism, propose a mechanism for cell trafficking through the placenta and discuss the different models and techniques available for its analysis. Likewise, we aim to generate interest in the use of ex vivo placenta perfusion to investigate microchimerism in physiological and pathological settings.
Collapse
Affiliation(s)
| | - Leonie Aengenheister
- Laboratory for Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Astrid Schmidt
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Udo R Markert
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany.
| | - Tina Buerki-Thurnherr
- Laboratory for Particles-Biology Interactions, Empa, Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | | |
Collapse
|
25
|
Koehn LM, Huang Y, Habgood MD, Nie S, Chiou SY, Banati RB, Dziegielewska KM, Saunders NR. Efflux transporters in rat placenta and developing brain: transcriptomic and functional response to paracetamol. Sci Rep 2021; 11:19878. [PMID: 34615937 PMCID: PMC8494792 DOI: 10.1038/s41598-021-99139-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 09/08/2021] [Indexed: 11/29/2022] Open
Abstract
Adenosine triphosphate binding cassette (ABC) transporters transfer lipid-soluble molecules across cellular interfaces either directly or after enzymatic metabolism. RNAseq analysis identified transcripts for ABC transporters and enzymes in rat E19, P5 and adult brain and choroid plexus and E19 placenta. Their functional capacity to efflux small molecules was studied by quantitative analysis of paracetamol (acetaminophen) and its metabolites using liquid scintillation counting, autoradiography and ultra-performance liquid chromatography coupled with tandem mass spectrometry (UPLC-MS/MS). Animals were treated acutely (30 min) and chronically (5 days, twice daily) with paracetamol (15 mg/kg) to investigate ability of brain and placenta barriers to regulate ABC transport functionality during extended treatment. Results indicated that transcripts of many efflux-associated ABC transporters were higher in adult brain and choroid plexus than at earlier ages. Chronic treatment upregulated certain transcripts only in adult brain and altered concentrations of paracetamol metabolites in circulation of pregnant dams. Combination of changes to metabolites and transport system transcripts may explain observed changes in paracetamol entry into adult and fetal brains. Analysis of lower paracetamol dosing (3.75 mg/kg) indicated dose-dependent changes in paracetamol metabolism. Transcripts of ABC transporters and enzymes at key barriers responsible for molecular transport into the developing brain showed alterations in paracetamol pharmacokinetics in pregnancy following different treatment regimens.
Collapse
Affiliation(s)
- L M Koehn
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Y Huang
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - M D Habgood
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - S Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3010, Australia
| | - S Y Chiou
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia
| | - R B Banati
- ANSTO - Australia's Nuclear Science and Technology Organisation, New Illawarra Rd, Lucas Heights, NSW, 2234, Australia.,University of Sydney, Camperdown, Sydney, Australia
| | - K M Dziegielewska
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia.,Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - N R Saunders
- Department of Biochemistry & Pharmacology, University of Melbourne, Parkville, VIC, 3010, Australia. .,Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
26
|
Ikumi NM, Pillay K, Tilburgs T, Malaba TR, Dzanibe S, Enninga EAL, Chakraborty R, Lamorde M, Myer L, Khoo S, Jaspan HB, Gray CM. T cell Homeostatic Imbalance in Placentae from Women with HIV in the absence of Vertical Transmission. J Infect Dis 2021; 224:S670-S682. [PMID: 33880544 DOI: 10.1093/infdis/jiab192] [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: 11/14/2022] Open
Abstract
BACKGROUND Implementation of universal antiretroviral therapy (ART) has significantly lowered vertical transmission rates but has also increased numbers of HIV-exposed uninfected children (HEU), who remain vulnerable to morbidities. Here, we investigated whether T cell alterations in the placenta contribute to altered immune status in HEU. METHODS We analyzed T cells from term placentae decidua and villous tissue and paired cord blood from pregnant women with HIV (PWH) who initiated ART late in pregnancy (n=21) with pregnant women not living with HIV (PWNH) (n=9). RESULTS Placentae from PWH showed inverted CD4:CD8 ratios and higher proportions of tissue resident CD8+ T cells in villous tissue relative to control placentae. CD8+ T cells in the fetal capillaries, which were of fetal origin, positively correlated with maternal plasma viraemia prior to ART initiation, implying that imbalanced T cells persisted throughout pregnancy. Additionally, the expanded memory differentiation of CD8+ T cells was confined to the fetal placental compartment and cord blood but was not observed in the maternal decidua. CONCLUSIONS T cell homeostatic imbalance in the blood circulation of PWH is reflected in the placenta. The placenta may be a causal link between HIV-induced maternal immune changes during gestation and altered immunity in newborn infants in the absence of vertical transmission.
Collapse
Affiliation(s)
- Nadia M Ikumi
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Komala Pillay
- National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa.,Division of Anatomical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Tamara Tilburgs
- Division of Immunobiology, Center for Inflammation and Tolerance, Cincinnati Children's Hospital, Cincinnati OH 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati OH 45229, USA
| | - Thokozile R Malaba
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Sonwabile Dzanibe
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | | | - Rana Chakraborty
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN.,Department of Pediatric and Adolescent Medicine, Mayo Clinic College of Medicine and Science, Minnesota, USA.,Department of Immunology, Mayo Clinic, Rochester, MN
| | - Mohammed Lamorde
- Infectious Diseases Institute, College of Health Sciences, Makerere University, Kampala, Uganda
| | - Landon Myer
- Division of Epidemiology and Biostatistics, School of Public Health and Family Medicine, University of Cape Town, Cape Town, South Africa
| | - Saye Khoo
- Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, UK.,Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Heather B Jaspan
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa
| | - Clive M Gray
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, South Africa.,National Health Laboratory Services, Groote Schuur Hospital, Cape Town, South Africa.,Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | |
Collapse
|
27
|
Alekar A. The outcomes of fetal cell microchimerism in the mother. BIOMEDICAL RESEARCH JOURNAL 2021. [DOI: 10.4103/bmrj.bmrj_9_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
28
|
Lessons from the Embryo: an Unrejected Transplant and a Benign Tumor. Stem Cell Rev Rep 2020; 17:850-861. [PMID: 33225425 DOI: 10.1007/s12015-020-10088-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2020] [Indexed: 10/22/2022]
Abstract
Embryogenesis is regarded the 'miracle of life', yet numerous aspects of this process are not fully understood. As the embryo grows in the mother's womb, immune components, stem cells and microenvironmental cues cooperate among others to promote embryonic development. Evidently, these key players are frequently associated with transplantation failure and tumor growth. While the fields of transplantation and cancer biology do not overlap, both can be viewed from the perspective of an embryo. As an 'unrejected transplant' and a 'benign tumor', lessons from embryonic development may reveal features of transplants and tumors that have been overlooked. Therefore, eavesdropping at these natural complex events during pregnancy may inspire more durable approaches to arrest transplant rejection or cancer progression.
Collapse
|
29
|
Koehn LM, Huang Y, Habgood MD, Kysenius K, Crouch PJ, Dziegielewska KM, Saunders NR. Effects of paracetamol (acetaminophen) on gene expression and permeability properties of the rat placenta and fetal brain. F1000Res 2020; 9:573. [PMID: 32934805 PMCID: PMC7477648 DOI: 10.12688/f1000research.24119.1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2020] [Indexed: 03/30/2024] Open
Abstract
Background: Paracetamol (acetaminophen) is widely used in pregnancy and generally regarded as "safe" by regulatory authorities. Methods: Clinically relevant doses of paracetamol were administered intraperitoneally to pregnant rats twice daily from embryonic day E15 to 19 (chronic) or as a single dose at E19 (acute). Control samples were from un-treated age-matched animals. At E19, rats were anaesthetised, administered a final paracetamol dose, uteruses were opened and fetuses exposed for sample collection. For RNA sequencing, placentas and fetal brains were removed and flash frozen. Fetal and maternal plasma and cerebrospinal fluid were assayed for ⍺-fetoprotein and interleukin 1β (IL1β). Brains were fixed and examined (immunohistochemistry) for plasma protein distribution. Placental permeability to a small molecule ( 14C-sucrose) was tested by injection into either mother or individual fetuses; fetal and maternal blood was sampled at regular intervals to 90 minutes. Results: RNA sequencing revealed a large number of genes up- or down-regulated in placentas from acutely or chronically treated animals compared to controls. Most notable was down-regulation of three acute phase plasma proteins (⍺-fetoprotein, transferrin, transthyretin) in acute and especially chronic experiments and marked up-regulation of immune-related genes, particularly cytokines, again especially in chronically treated dams. IL1β increased in plasma of most fetuses from treated dams but to variable levels and no IL1β was detectable in plasma of control fetuses or any of the dams. Increased placental permeability appeared to be only from fetus to mother for both 14C-sucrose and ⍺-fetoprotein, but not in the reverse direction. In the fetal brain, gene regulatory changes were less prominent than in the placenta of treated fetuses and did not involve inflammatory-related genes; there was no evidence of increased blood-brain barrier permeability. Conclusion: Results suggest that paracetamol may induce an immune-inflammatory-like response in placenta and more caution should be exercised in use of paracetamol in pregnancy.
Collapse
Affiliation(s)
- Liam M. Koehn
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yifan Huang
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mark D Habgood
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Kai Kysenius
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Peter J. Crouch
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | - Norman R Saunders
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
30
|
Koehn LM, Huang Y, Habgood MD, Kysenius K, Crouch PJ, Dziegielewska KM, Saunders NR. Effects of paracetamol (acetaminophen) on gene expression and permeability properties of the rat placenta and fetal brain. F1000Res 2020; 9:573. [PMID: 32934805 PMCID: PMC7477648 DOI: 10.12688/f1000research.24119.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2020] [Indexed: 01/10/2023] Open
Abstract
Background: Paracetamol (acetaminophen) is widely used in pregnancy and generally regarded as "safe" by regulatory authorities. Methods: Clinically relevant doses of paracetamol were administered intraperitoneally to pregnant rats twice daily from embryonic day E15 to 19 (chronic) or as a single dose at E19 (acute). Control samples were from un-treated age-matched animals. At E19, rats were anaesthetised, administered a final paracetamol dose, uteruses were opened and fetuses exposed for sample collection. For RNA sequencing, placentas and fetal brains were removed and flash frozen. Fetal and maternal plasma and cerebrospinal fluid were assayed for α-fetoprotein and interleukin 1β (IL1β). Brains were fixed and examined (immunohistochemistry) for plasma protein distribution. Placental permeability to a small molecule ( 14C-sucrose) was tested by injection into either mother or individual fetuses; fetal and maternal blood was sampled at regular intervals to 90 minutes. Results: RNA sequencing revealed a large number of genes up- or down-regulated in placentas from acutely or chronically treated animals compared to controls. Most notable was down-regulation of three acute phase plasma proteins (α-fetoprotein, transferrin, transthyretin) in acute and especially chronic experiments and marked up-regulation of immune-related genes, particularly cytokines, again especially in chronically treated dams. IL1β increased in plasma of most fetuses from treated dams but to variable levels and no IL1β was detectable in plasma of control fetuses or any of the dams. Increased placental permeability appeared to be only from fetus to mother for both 14C-sucrose and α-fetoprotein, but not in the reverse direction. In the fetal brain, gene regulatory changes were less prominent than in the placenta of treated fetuses and did not involve inflammatory-related genes; there was no evidence of increased blood-brain barrier permeability. Conclusion: Results suggest that paracetamol may induce an immune-inflammatory-like response in placenta and more caution should be exercised in use of paracetamol in pregnancy.
Collapse
Affiliation(s)
- Liam M. Koehn
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Yifan Huang
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mark D Habgood
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Kai Kysenius
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Peter J. Crouch
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | - Norman R Saunders
- Pharmacology & Therapeutics, University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
31
|
Saleh M, Taher M, Sohrabpour AA, Vaezi AA, Nasiri Toosi M, Kavianpour M, Ghazvinian Z, Abdolahi S, Verdi J. Perspective of placenta derived mesenchymal stem cells in acute liver failure. Cell Biosci 2020; 10:71. [PMID: 32483484 PMCID: PMC7245988 DOI: 10.1186/s13578-020-00433-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023] Open
Abstract
Acute Liver failure (ALF) is a life-threatening disease and is determined by coagulopathy (with INR ≥ 1.5) and hepatic encephalopathy as a result of severe liver injury in patients without preexisting liver disease. Since there are problems with liver transplantation including lack of donors, use of immunosuppressive drugs, and high costs of this process, new therapeutic approaches alongside current treatments are needed. The placenta is a tissue that is normally discarded after childbirth. On the other hand, human placenta is a rich source of mesenchymal stem cells (MSCs), which is easily available, without moral problems, and its derived cells are less affected by age and environmental factors. Therefore, placenta-derived mesenchymal stem cells (PD-MSCs) can be considered as an allogeneic source for liver disease. Considering the studies on MSCs and their effects on various diseases, it can be stated that MSCs are among the most important agents to be used for novel future therapies of liver diseases. In this paper, we will investigate the effects of mesenchymal stem cells through migration and immigration to the site of injury, cell-to-cell contact, immunomodulatory effects, and secretory factors in ALF.
Collapse
Affiliation(s)
- Mahshid Saleh
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taher
- 2Gastroenterology and Hepatology, Tehran University of Medical Sciences, Imam Hospital Complex, Tehran, Iran
| | - Amir Ali Sohrabpour
- 3Gastroenterology and Hepatology, School of Medicine Shariati Hospital, Tehran University of Medical Science, Tehran, Iran
| | - Amir Abbas Vaezi
- 4Department of Internal Medicine, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohsen Nasiri Toosi
- 5Internal Medicine, School of Medicine Liver Transplantation Research Center Imam, Khomeini Hospital Tehran University of Medical Sciences, Tehran, Iran
| | - Maria Kavianpour
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Ghazvinian
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrokh Abdolahi
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Verdi
- 1Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Moser G, Guettler J, Forstner D, Gauster M. Maternal Platelets—Friend or Foe of the Human Placenta? Int J Mol Sci 2019; 20:ijms20225639. [PMID: 31718032 PMCID: PMC6888633 DOI: 10.3390/ijms20225639] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/07/2019] [Accepted: 11/08/2019] [Indexed: 12/25/2022] Open
Abstract
Human pregnancy relies on hemochorial placentation, including implantation of the blastocyst and deep invasion of fetal trophoblast cells into maternal uterine blood vessels, enabling direct contact of maternal blood with placental villi. Hemochorial placentation requires fast and reliable hemostasis to guarantee survival of the mother, but also for the neonates. During human pregnancy, maternal platelet count decreases gradually from first, to second, and third trimester. In addition to hemodilution, accelerated platelet sequestration and consumption in the placental circulation may contribute to a decline of platelet count throughout gestation. Local stasis, turbulences, or damage of the syncytiotrophoblast layer can activate maternal platelets within the placental intervillous space and result in formation of fibrin-type fibrinoid. Perivillous fibrinoid is a regular constituent of the normal placenta which is considered to be an important regulator of intervillous hemodynamics, as well as having a role in shaping the developing villous trees. However, exaggerated activation of platelets at the maternal-fetal interface can provoke inflammasome activation in the placental trophoblast, and enhance formation of circulating platelet-monocyte aggregates, resulting in sterile inflammation of the placenta and a systemic inflammatory response in the mother. Hence, the degree of activation determines whether maternal platelets are a friend or foe of the human placenta. Exaggerated activation of maternal platelets can either directly cause or propagate the disease process in placenta-associated pregnancy pathologies, such as preeclampsia.
Collapse
|
33
|
Liu J, Mosavati B, Oleinikov AV, Du E. Biosensors for Detection of Human Placental Pathologies: A Review of Emerging Technologies and Current Trends. Transl Res 2019; 213:23-49. [PMID: 31170377 PMCID: PMC6783355 DOI: 10.1016/j.trsl.2019.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 02/06/2023]
Abstract
Substantial growth in the biosensor research has enabled novel, sensitive and point-of-care diagnosis of human diseases in the last decade. This paper presents an overview of the research in the field of biosensors that can potentially predict and diagnosis of common placental pathologies. A survey of biomarkers in maternal circulation and their characterization methods is presented, including markers of oxidative stress, angiogenic factors, placental debris, and inflammatory biomarkers that are associated with various pathophysiological processes in the context of pregnancy complications. Novel biosensors enabled by microfluidics technology and nanomaterials is then reviewed. Representative designs of plasmonic and electrochemical biosensors for highly sensitive and multiplexed detection of biomarkers, as well as on-chip sample preparation and sensing for automatic biomarker detection are illustrated. New trends in organ-on-a-chip based placental disease models are highlighted to illustrate the capability of these in vitro disease models in better understanding the complex pathophysiological processes, including mass transfer across the placental barrier, oxidative stress, inflammation, and malaria infection. Biosensor technologies that can be potentially embedded in the placental models for real time, label-free monitoring of these processes and events are suggested. Merger of cell culture in microfluidics and biosensing can provide significant potential for new developments in advanced placental models, and tools for diagnosis, drug screening and efficacy testing.
Collapse
Affiliation(s)
- Jia Liu
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida
| | - Babak Mosavati
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida
| | - Andrew V Oleinikov
- Charles E. Schmidt College of Medicine, Department of Biomedical Science, Florida Atlantic University, Boca Raton, Florida
| | - E Du
- College of Engineering and Computer Science, Department of Ocean and Mechanical Engineering, Florida Atlantic University, Boca Raton, Florida; Charles E. Schmidt College of Science, Department of Biological Sciences, Florida Atlantic University, Boca Raton, Florida.
| |
Collapse
|
34
|
Abstract
Malignant blood disorders depend on heritable susceptibility genes and occur in familial aggregations. We suggest a model of transgenerational segregation of the susceptibility genes based on the study of malignant blood disorders in Norwegian and Danish families with unrelated parents, and in the inbred Faroese population with related parents. This model, consisting of parental genomic imprinting and mother-son microchimerism, can explain the male predominance in most of the diseases, the predominance of affected parent-offspring when parents are not related, and the different modes of segregation in males and females. The model displays a specific pattern in the distribution of affected relatives for each diagnosis, viz. a characteristic distribution in the pedigrees of family members with malignant blood disorder related to the proband. Three such patterns, each reflecting a specific transgenerational passage, were identified: (1) alterations in the number of affected relatives in paternal lines alone, e.g. in patterns for probands with multiple myeloma; (2) alterations in the number of affected relatives in both paternal and maternal lines for probands with chronic lymphocytic leukemia; and (3) no alterations in the numbers of male and female affected relatives in the parental lines, e.g. for probands with some types of malignant lymphoma.
Collapse
|
35
|
Gokhale S, Gokhale S. Transfusing maternal blood to her newborn baby-irrespective of ABO mismatch. J Matern Fetal Neonatal Med 2019; 33:1593-1606. [PMID: 30686061 DOI: 10.1080/14767058.2018.1525355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Background: Though blood transfusions are the common procedures in pediatric patients, transfusion reactions are rare in children. Though in adults, uncross-matched ABO group-specific blood is used in emergencies, there are no such reports in neonates and children. There are stray case reports about transfusing maternal blood for her baby and maternal blood is de facto compatible regardless of an ABO mismatchObjective: Confirming our previous hypothesis that maternal blood is compatible with her baby's blood; and maternal blood can be used for transfusion in her newborn baby irrespective of ABO match/ mismatch.Design: Prospective interventional study.Setting and Participants: Fifty-one mother-baby pairs were recruited attending Pediatric Unit of our Community Hospital from 15 July 2013 to 13 July 2015. After obtaining consent from the parents, all the required lab tests were done. Since all lab reports were favourable; these babies qualified for transfusion of maternal blood.Interventions: Fifty-one sick newborns were transfused fresh whole maternal blood as a part of treatment; irrespective of mother-baby ABO match or mismatch.Results: All babies tolerated maternal blood well and showed significant and rapid improvement. Minimum period of observation was from a minimum of 32 to a maximum of 56 months. All the babies showed good growth and development.Conclusion: By observing a particular protocol and procedural techniques, mother's blood may be used for transfusion in her own baby in neonatal period, irrespective of ABO mismatch.Significance: This is probably the largest series in world literature of 51 newborns being transfused maternal blood either ABO match or mismatch.
Collapse
Affiliation(s)
- Sanjay Gokhale
- Department of Pediatrics, Rajhans Hospital, Mumbai, India
| | - Sankalp Gokhale
- Department of Neurology [Medicine], Duke University, Durham, NC, USA
| |
Collapse
|
36
|
Abstract
Microchimerism is the presence of cells from one individual in another genetically distinct individual. Pregnancy is the main cause of natural microchimerism through transplacental bi-directional cell trafficking between mother and fetus. In addition to a variety of cell-free substances, it is now well-recognized that some cells are also exchanged in pregnancy. Furthermore, it is now known that microchimerism persists decades later both in mother and in her progeny. The consequences of pregnancy-related microchimerism are under active investigation. However, many authors have suggested a close relationship linking fetal microchimerism and the development of autoimmune diseases. Fetal microchimerism is emerging as a potential contributing factor in certain diseases, including cancer. Parallel studies in animal and human pregnancy suggest that microchimeric fetal cells play a role in wound healing. Role of these microchimeric cells in human health and disease is discussed here.
Collapse
Affiliation(s)
- Sandhya Shrivastava
- Department of Oral Pathology and Microbiology, Hi-Tech Dental College and Research Institute, Bhubneshwar, Odisha, India
| | - Rupali Naik
- Dental Surgeon, Rural Hospital, Devrukh, Ratnagiri, Maharashtra, India
| | - Hema Suryawanshi
- Department of Oral Pathology and Microbiology, Chhattisgarh Dental College and Research Institute, Rajnandgaon, Chhattisgarh, India
| | - Neha Gupta
- Research Scientist and Consultant Stemgenn Therapeutics, New Delhi, India
| |
Collapse
|
37
|
Kajbafzadeh AM, Sabetkish S, Sabetkish N. The role of fetal-maternal microchimerism as a natural-born healer in integrity improvement of maternal damaged kidney. Int Braz J Urol 2018; 44:608-616. [PMID: 29211403 PMCID: PMC5996814 DOI: 10.1590/s1677-5538.ibju.2017.0324] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/04/2017] [Indexed: 12/11/2022] Open
Abstract
PURPOSE To identify the fetal stem cell (FSC) response to maternal renal injury with emphasis on renal integrity improvement and Y chromosome detection in damaged maternal kidney. MATERIALS AND METHODS Eight non-green fluorescent protein (GFP) transgenic Sprague- Dawley rats were mated with GFP-positive transgenic male rats. Renal damage was induced on the right kidney at gestational day 11. The same procedure was performed in eight non-pregnant rats as control group. Three months after delivery, right nephrectomy was performed in order to evaluate the injured kidney. The fresh perfused kidneys were stained with anti-GFP antibody. Polymerase chain reaction (PCR) assay was also performed for the Y chromosome detection. Cell culture was performed to detect the GFP-positive cells. Technetium-99m-DMSA renal scan and single-photon emission computed tomography (SPECT) were performed after renal damage induction and 3 months later to evaluate the improvement of renal integrity. RESULTS The presence of FSCs was confirmed by immune histochemical staining as well as immunofluorescent imaging of the damaged part. Gradient PCR of female rat purified DNA demonstrated the presence of Y-chromosome in the damaged maternal kidney. Moreover, the culture of kidney cells showed GPF- positive cells by immunofluorescence microscopy. The acute renal scar was repaired and the integrity of damaged kidney reached to near normal levels in experimental group as shown in DMSA scan. However, no significant improvement was observed in control group. CONCLUSION FSC seems to be the main mechanism in repairing of the maternal renal injury during pregnancy as indicated by Y chromosome and GFP-positive cells in the sub-cultured medium.
Collapse
Affiliation(s)
- Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shabnam Sabetkish
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Sabetkish
- Pediatric Urology and Regenerative Medicine Research Center, Section of Tissue Engineering and Stem Cells Therapy, Children's Hospital Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Ståhlberg A, El-Heliebi A, Sedlmayr P, Kroneis T. Unravelling the biological secrets of microchimerism by single-cell analysis. Brief Funct Genomics 2018; 17:255-264. [PMID: 29028900 PMCID: PMC6063264 DOI: 10.1093/bfgp/elx027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The presence of microchimeric cells is known for >100 years and well documented since decades. Earlier, microchimeric cells were mainly used for cell-based non-invasive prenatal diagnostics during early pregnancy. Microchimeric cells are also present beyond delivery and are associated to various autoimmune diseases, tissue repair, cancer and immune tolerance. All these findings were based on low complexity studies and occasionally accompanied by artefacts not allowing the biological functions of microchimerism to be determined. However, with the recent developments in single-cell analysis, new means to identify and characterize microchimeric cells are available. Cell labelling techniques in combination with single-cell analysis provide a new toolbox to decipher the biology of microchimeric cells at molecular and cellular level. In this review, we discuss how recent developments in single-cell analysis can be applied to determine the role and function of microchimeric cells.
Collapse
Affiliation(s)
- Anders Ståhlberg
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
| | - Amin El-Heliebi
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Peter Sedlmayr
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| | - Thomas Kroneis
- Sahlgrenska Cancer Center, Department of Pathology and Genetics, Institute of Biomedicine, Sahlgrenska Academy at University of Gothenburg, Medicinaregatan 1F, Gothenburg, Sweden
- Institute of Cell Biology, Histology & Embryology, Medical University of Graz, Harrachgasse 21, Graz, Austria
| |
Collapse
|
39
|
van Otterdijk SD, Binder AM, Michels KB. Locus-specific DNA methylation in the placenta is associated with levels of pro-inflammatory proteins in cord blood and they are both independently affected by maternal smoking during pregnancy. Epigenetics 2017; 12:875-885. [PMID: 28820654 DOI: 10.1080/15592294.2017.1361592] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
We investigated the impact of maternal smoking during pregnancy on placental DNA methylation and how this may mediate the association between maternal smoking and pro-inflammatory proteins in cord blood. The study population consisted of 27 individuals exposed to maternal smoking throughout pregnancy, 32 individuals exposed during a proportion of the pregnancy, and 61 unexposed individuals. Methylation of 11 regions within 6 genes in placenta tissue was assessed by pyrosequencing. Levels of 7 pro-inflammatory proteins in cord blood were assessed by electrochemiluminescence. Differential methylation was observed in the CYP1A1 promoter and AHRR gene body regions between women who smoked throughout pregnancy and non-smokers on the fetal-side of the placenta and in the GFI1 promoter between women who quit smoking while pregnant and non-smokers on the maternal-side of the placenta. Maternal smoking resulted in elevated levels of IL-8 protein in cord blood, which was not mediated by DNA methylation of our candidate regions at either the maternal or the fetal side of the placenta. Placental DNA methylation was associated with levels of inflammatory proteins in cord blood. Our observations suggest that maternal smoking during pregnancy affects both placental DNA methylation and the neonate's immune response.
Collapse
Affiliation(s)
- Sanne D van Otterdijk
- a Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center , University of Freiburg , Freiburg , Germany.,b Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics, Gynecology and Reproductive Biology , Brigham and Women's Hospital, Harvard Medical School , Boston , MA
| | - Alexandra M Binder
- b Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics, Gynecology and Reproductive Biology , Brigham and Women's Hospital, Harvard Medical School , Boston , MA.,c Department of Epidemiology , Harvard School of Public Health , Boston , MA , USA
| | - Karin B Michels
- a Institute for Prevention and Cancer Epidemiology, Faculty of Medicine and Medical Center , University of Freiburg , Freiburg , Germany.,b Obstetrics and Gynecology Epidemiology Center, Department of Obstetrics, Gynecology and Reproductive Biology , Brigham and Women's Hospital, Harvard Medical School , Boston , MA.,c Department of Epidemiology , Harvard School of Public Health , Boston , MA , USA
| |
Collapse
|
40
|
Riccardo F, Réal A, Voena C, Chiarle R, Cavallo F, Barutello G. Maternal Immunization: New Perspectives on Its Application Against Non-Infectious Related Diseases in Newborns. Vaccines (Basel) 2017; 5:E20. [PMID: 28763018 PMCID: PMC5620551 DOI: 10.3390/vaccines5030020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 07/24/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
The continuous evolution in preventive medicine has anointed vaccination a versatile, human-health improving tool, which has led to a steady decline in deaths in the developing world. Maternal immunization represents an incisive step forward for the field of vaccination as it provides protection against various life-threatening diseases in pregnant women and their children. A number of studies to improve prevention rates and expand protection against the largest possible number of infections are still in progress. The complex unicity of the mother-infant interaction, both during and after pregnancy and which involves immune system cells and molecules, is an able partner in the success of maternal immunization, as intended thus far. Interestingly, new studies have shed light on the versatility of maternal immunization in protecting infants from non-infectious related diseases, such as allergy, asthma and congenital metabolic disorders. However, barely any attempt at applying maternal immunization to the prevention of childhood cancer has been made. The most promising study reported in this new field is a recent proof of concept on the efficacy of maternal immunization in protecting cancer-prone offspring against mammary tumor progression. New investigations into the possibility of exploiting maternal immunization to prevent the onset and/or progression of neuroblastoma, one of the most common childhood malignancies, are therefore justified. Maternal immunization is presented in a new guise in this review. Attention will be focused on its versatility and potential applications in preventing tumor progression in neuroblastoma-prone offspring.
Collapse
Affiliation(s)
- Federica Riccardo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Aline Réal
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Claudia Voena
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health Sciences, Center for Experimental Research and Medical Studies, University of Torino, Torino 10126, Italy.
- Department of Pathology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Federica Cavallo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| | - Giuseppina Barutello
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino 10126, Italy.
| |
Collapse
|
41
|
|
42
|
Abstract
OBJECTIVE Zika virus infection during pregnancy is a cause of microcephaly and other fetal brain abnormalities. Reports indicate that the duration of detectable viral RNA in serum after symptom onset is brief. In a recent case report involving a severely affected fetus, Zika virus RNA was detected in maternal serum 10 weeks after symptom onset, longer than the duration of RNA detection in serum previously reported. This report summarizes the clinical and laboratory characteristics of pregnant women with prolonged detection of Zika virus RNA in serum that were reported to the U.S. Zika Pregnancy Registry. METHODS Data were obtained from the U.S. Zika Pregnancy Registry, an enhanced surveillance system of pregnant women with laboratory evidence of confirmed or possible Zika virus infection. For this case series, we defined prolonged detection of Zika virus RNA as Zika virus RNA detection in serum by real-time reverse transcription-polymerase chain reaction (RT-PCR) 14 or more days after symptom onset or, for women not reporting signs or symptoms consistent with Zika virus disease (asymptomatic), 21 or more days after last possible exposure to Zika virus. RESULTS Prolonged Zika virus RNA detection in serum was identified in four symptomatic pregnant women up to 46 days after symptom onset and in one asymptomatic pregnant woman 53 days postexposure. Among the five pregnancies, one pregnancy had evidence of fetal Zika virus infection confirmed by histopathologic examination of fetal tissue, three pregnancies resulted in live births of apparently healthy neonates with no reported abnormalities, and one pregnancy is ongoing. CONCLUSION Zika virus RNA was detected in the serum of five pregnant women beyond the previously estimated timeframe. Additional real-time RT-PCR testing of pregnant women might provide more data about prolonged detection of Zika virus RNA and the possible diagnostic, epidemiologic, and clinical implications for pregnant women.
Collapse
|
43
|
Abstract
ABSTRACT
The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Collapse
|
44
|
Stefanovic V. Fetomaternal hemorrhage complicated pregnancy: risks, identification, and management. Curr Opin Obstet Gynecol 2016; 28:86-94. [PMID: 26866844 DOI: 10.1097/gco.0000000000000248] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW This article aims not only to review recent literature about the clinical features of massive fetomaternal hemorrhage (FMH) and identification of risk factors, but also to alert obstetricians and pediatricians to this underdiagnosed and underestimated severe obstetrical issue. In addition, a simplified flow chart for the antenatal management of suspected FMH is proposed. RECENT FINDINGS Improvements in obstetrical and neonatal care have decreased perinatal morbidity and mortality and the rate of stillbirth. Unfortunately, because of the nonspecific signs on presentation, adverse outcome associated with massive FMH has not followed this trend and still has devastating consequences. As even the definition varies among publications and there is lack of universal screening, the real nature still remains obscure. Improvements in the diagnosis of fetal anemia, laboratory and intrauterine transfusion techniques, and the implementation of prenatal and postnatal neuroprotection give some hope for the better outcome in the most severe cases. Unfortunately, obstetricians' awareness of the massive FMH remains still at an unacceptably low level. SUMMARY There is an urgent need for the internationally accepted definition, standardized pregnancy management protocol, and structured follow-up of neonates from such pregnancies. We suggest the international registry of massive FMH cases.
Collapse
Affiliation(s)
- Vedran Stefanovic
- Department of Obstetrics and Gynecology, Fetomaternal Medical Center, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
45
|
Curtis BR. Recent progress in understanding the pathogenesis of fetal and neonatal alloimmune thrombocytopenia. Br J Haematol 2015; 171:671-82. [PMID: 26344048 DOI: 10.1111/bjh.13639] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fetal and neonatal alloimmune thrombocytopenia (FNAIT) occurs in c. 1 in 1000 births and is caused by maternal antibodies against human platelet alloantigens that bind incompatible fetal platelets and promote their clearance from the circulation. Affected infants can experience bleeding, bruising and, in severe cases, intracranial haemorrhage and even death. As maternal screening is not routinely performed, and first pregnancies can be affected, most cases are diagnosed at delivery of a first affected pregnancy. Unlike its erythrocyte counterpart, Haemolytic Disease of the Fetus and Newborn, there is no prophylactic treatment for FNAIT. This report will review recent advances made in understanding the pathogenesis of FNAIT: the platelet alloantigens involved, maternal exposure and sensitization to fetal platelet antigens, properties of platelet Immunoglobulin G antibodies, maternal-fetal antibody transport mechanisms and efforts to develop an effective FNAIT prophylaxis.
Collapse
Affiliation(s)
- Brian R Curtis
- Platelet & Neutrophil Immunology Laboratory and Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
46
|
Maternal microchimerism: lessons learned from murine models. J Reprod Immunol 2015; 108:12-25. [DOI: 10.1016/j.jri.2014.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 12/04/2014] [Accepted: 12/14/2014] [Indexed: 11/20/2022]
|
47
|
Pabón MM, Ji X, Fernandez JW, Borlongan CV. Gender-linked stem cell alterations in stroke and postpartum depression. CNS Neurosci Ther 2015; 21:348-56. [PMID: 25399760 PMCID: PMC4376590 DOI: 10.1111/cns.12339] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 09/19/2014] [Accepted: 09/23/2014] [Indexed: 12/15/2022] Open
Abstract
Stroke is a significant unmet clinical need. The current stroke treatment of tissue plasminogen activator is limited to the very acute 4.5 h after disease onset which benefits only less than 3% of ischemic stroke patients. Our overarching hypothesis advances the notion that gender, which has been established as a comorbidity factor of stroke, plays a key role in regenerative medicine, in particular stem cell therapy. We hypothesize that gender is a key factor in culture-induced stemness of adult stem cells. Our goal is to provide new evidence supporting gender effects on stroke and stem cells for the purpose of enhancing our understanding of the pathophysiology of the disease and developing novel stem cell-based therapeutics targeting gender-relevant stress hormones as manifested in a stroke-postpartum depression paradigm.
Collapse
Affiliation(s)
- Mibel M. Pabón
- Department of Neurosurgery and Brain RepairMorsani College of MedicineUniversity of South Florida College of MedicineTampaFLUSA
| | - Xun‐Ming Ji
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| | - Jamie Winderbaum Fernandez
- Department of Psychiatry and Behavioral NeurosciencesMorsani College of MedicineUniversity of South Florida College of MedicineTampaFLUSA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain RepairMorsani College of MedicineUniversity of South Florida College of MedicineTampaFLUSA
- Department of NeurosurgeryXuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
48
|
Bella L, Zona S, Nestal de Moraes G, Lam EWF. FOXM1: A key oncofoetal transcription factor in health and disease. Semin Cancer Biol 2014; 29:32-9. [PMID: 25068996 DOI: 10.1016/j.semcancer.2014.07.008] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 07/17/2014] [Indexed: 12/25/2022]
Abstract
Forkhead Box M1 (FOXM1) is a bona fide oncofoetal transcription factor, which orchestrates complex temporal and spatial gene expression throughout embryonic and foetal development as well as during adult tissue homeostasis and repair. Controlled FOXM1 expression and activity provides a balanced transcriptional programme to ensure proper growth and maturation during embryogenesis and foetal development as well as to manage appropriate homeostasis and repair of adult tissues. Conversely, deregulated FOXM1 upregulation likely affects cell migration, invasion, angiogenesis, stem cell renewal, DNA damage repair and cellular senescence, which impact tumour initiation, progression, metastasis, angiogenesis and drug resistance. A thorough understanding of the regulation and role of FOXM1 in health and in cancer should contribute to the development of better diagnostics and treatments for cancer as well as congenital disorders and other developmental diseases.
Collapse
Affiliation(s)
- Laura Bella
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London W12 0NN, United Kingdom
| | - Stefania Zona
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London W12 0NN, United Kingdom
| | - Gabriela Nestal de Moraes
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London W12 0NN, United Kingdom
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM), Imperial College London, Hammersmith Hospital Campus, London W12 0NN, United Kingdom.
| |
Collapse
|
49
|
Shirasaka T, Hashimoto E, Ukai W, Yoshinaga T, Ishii T, Tateno M, Saito T. Stem cell therapy: social recognition recovery in a FASD model. Transl Psychiatry 2012; 2:e188. [PMID: 23149452 PMCID: PMC3565770 DOI: 10.1038/tp.2012.111] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
To better understand the cellular pathogenetic mechanisms of fetal alcohol spectrum disorder (FASD) and the therapeutic benefit of stem cell treatment, we exposed pregnant rats to ethanol followed by intravenous administration of neural stem cells (NSCs) complexed with atelocollagen to the new born rats and studied recovery of GABAergic interneuron numbers and synaptic protein density in the anterior cingulate cortex, hippocampus and amygdala. Prenatal ethanol exposure reduced both parvalbumin-positive phenotype of GABAergic interneurons and postsynaptic density protein 95 levels in these areas. Intravenous NSC treatment reversed these reductions. Furthermore, treatment with NSCs reversed impaired memory/cognitive function and social interaction behavior. These experiments underscore an important role for synaptic remodeling and GABAergic interneuron genesis in the pathophysiology and treatment of FASD and highlight the therapeutic potential for intravenous NSC administration in FASD utilizing atelocollagen.
Collapse
Affiliation(s)
- T Shirasaka
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan.
| | - E Hashimoto
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| | - W Ukai
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| | - T Yoshinaga
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| | - T Ishii
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| | - M Tateno
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| | - T Saito
- Department of Neuropsychiatry, School of Medicine, Sapporo Medical University, Chuo-ku, Sapporo, Japan
| |
Collapse
|
50
|
Abstract
The development of the adaptive immune system has been studied in the mouse primarily because it is easier to access fetal tissues and because there exists a rich array of probes for analysis of various components of the immune system. While much has been learned from this exercise, it is also clear that different species show substantial temporal variation in the development of the immune system during early life. In mice, for instance, mature α/β T cells first appear in the periphery during the final stages of fetal gestation and only increase in number after birth (Friedberg and Weissman, 1974); in humans, on the other hand, the first mature α/β T cells are seen in peripheral tissues at 10-12 gestational weeks (g.w.) and are circulating in significant numbers by the end of the second trimester (Ceppellini et al., 1971; Haynes et al., 1988; Hayward and Ezer, 1974; Kay et al., 1970). Although the functional implications of these differences remain unclear, it is likely that there are significant biological consequences associated with the relatively early development of the peripheral adaptive immune system in humans, for example, with respect to the development of peripheral tolerance as well as to the response to antigens that might cross the placenta from the mother (e.g., cells bearing noninherited maternal alloantigens, infectious agents, food antigens, and the like). Here, we will review studies of immune system ontogeny in the mouse and in humans, and then focus on the possible functional roles of fetal T cell populations during development and later in life in humans.
Collapse
Affiliation(s)
- Jeff E Mold
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|