1
|
Nguyen BN, Britten-Jones AC, Bui BV, Walker LE, Titter P. Physiological and pathological changes to the eye and vision during and after pregnancy. Clin Exp Optom 2024:1-9. [PMID: 39374945 DOI: 10.1080/08164622.2024.2410031] [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: 03/29/2024] [Revised: 09/02/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024] Open
Abstract
Pregnancy introduces a multitude of changes in the body, including hormonal fluctuations and metabolic changes, which can lead to atypical ocular signs and symptoms. Ocular manifestations range from fluctuations in vision, to microstructural changes in the retina and choroid, to dry eye disease. This narrative review highlights the range of pregnancy-related effects on the eye and vision that are likely to present in the context of routine eyecare. Specifically, physiological ocular changes and pathological ocular changes that manifest for the first time, or are exacerbated, in uncomplicated pregnancy are discussed. The literature has evolved from simply noting differences in the eye between pregnant and non-pregnant groups, to refining knowledge of the proposed underlying pathophysiology with the advent of newer technologies in eyecare. A particular focus of this review is navigating when pregnancy changes in the eye occur or peak during the gestational period, and whether the changes are short-lived or might extend past pregnancy. While many pregnancy-associated changes are temporary and resolve post-partum, it is also recognised that some changes persist after pregnancy, with a notable absence of literature on ocular changes with loss or termination of pregnancy. Currently or previously pregnant women (or those planning to become pregnant), and other health professionals, should be educated about the importance of seeking eyecare before, during and after pregnancy.
Collapse
Affiliation(s)
- Bao N Nguyen
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | | | - Bang V Bui
- Department of Optometry and Vision Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Lorraine E Walker
- School of Nursing and Midwifery, Monash University, Clayton, Victoria, Australia
| | - Peta Titter
- School of Nursing, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
2
|
Nowotny HF, Tschaidse L, Auer MK, Reisch N. Prenatal and Pregnancy Management of Congenital Adrenal Hyperplasia. Clin Endocrinol (Oxf) 2024; 101:359-370. [PMID: 39387451 DOI: 10.1111/cen.15131] [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: 03/22/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024]
Abstract
Management of patients with congenital adrenal hyperplasia (CAH) poses challenges during pregnancy and prenatal stages, impacting fertility differently in men and women. Women with CAH experience menstrual irregularities due to androgen and glucocorticoid precursor interference with endometrial development and ovulation. Genital surgeries for virilization and urogenital anomalies further impact fertility and sexual function, leading to reduced heterosexual relationships among affected women. Fertility rates vary, with a lower prevalence of motherhood, primarily among those with classic CAH, necessitating optimized hormonal therapy for conception. Monitoring optimal disease control during pregnancy poses challenges due to hormonal fluctuations. Men with CAH often experience hypogonadotrophic hypogonadism and complications like testicular adrenal rest tissue, impacting fertility. Regular monitoring and intensified glucocorticoid therapy may restore spermatogenesis. Genetic counselling is vital to comprehend transmission risks and prenatal implications. Prenatal dexamethasone treatment in affected female fetuses prevents virilization but raises ethical and safety concerns, necessitating careful consideration and further research. The international "PREDICT" study aims to establish safer and more effective prenatal therapy in CAH, evaluating dosage, safety, and long-term effects.
Collapse
Affiliation(s)
| | - Lea Tschaidse
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias K Auer
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Nicole Reisch
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
3
|
Reisch N, Auchus RJ. Pregnancy in Congenital Adrenal Hyperplasia. Endocrinol Metab Clin North Am 2024; 53:391-407. [PMID: 39084815 DOI: 10.1016/j.ecl.2024.05.005] [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] [Indexed: 08/02/2024]
Abstract
Over the last several decades, children with all forms of classic congenital adrenal hyperplasia (CAH) are identified early and treated appropriately throughout childhood. As adults, women with CAH may desire to become mothers and their usual chronic therapy and disease control is often inadequate for conception. Subsequently, little data exist on their management during pregnancy. Pregnancy in women with various forms of CAH is possible with appropriate treatment. Achieving pregnancy is more complex than disease management during pregnancy.
Collapse
Affiliation(s)
- Nicole Reisch
- Department of Medicine IV, Institute for Endocrinology, Diabetology & Metabolism, Klinikum der Universität München, Ziemssenstraße 1, München 80336, Germany
| | - Richard J Auchus
- Department of Pharmacology, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, MSRB II, 5560A, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA; Department of Internal Medicine, Division of Metabolism, Endocrinology and Diabetes, University of Michigan, MSRB II, 5560A, 1150 West Medical Center Drive, Ann Arbor, MI 48109, USA.
| |
Collapse
|
4
|
Benzi JRDL, Tsang YP, Unadkat JD. The effect of pregnancy-related hormones on hepatic transporters: studies with premenopausal human hepatocytes. Front Pharmacol 2024; 15:1440010. [PMID: 39170705 PMCID: PMC11335556 DOI: 10.3389/fphar.2024.1440010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/18/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Pregnancy results in significant changes in drug pharmacokinetics (PK). While previous studies have elucidated the impact of pregnancy-related hormones (PRH) on mRNA or protein expression and activity of major hepatic metabolizing enzymes, their effect on hepatic drug transporters remains largely unexplored. Therefore, we investigated the effect of a cocktail of PRH on the mRNA expression and activity of hepatic transporters. Methods Plated human hepatocytes (PHH) from 3 premenopausal donors were incubated, in triplicate, for 72 h, with vehicle (DMSO < 0.01%), rifampin (10 μM; positive control) or a cocktail of PRH consisting of estrone, estradiol, estriol, estetrol, progesterone, cortisol, testosterone, oxytocin, and placental growth hormone. The PRH concentrations replicated 0.1×, 1×, or 10× of the plasma concentrations of these hormones observed during each of the three trimesters of pregnancy. After treatment, mRNA expression (quantified by qPCR) of hepatic influx and efflux transporters as well as the activity of influx transporters was quantified (uptake of a selective substrate ± corresponding transporter inhibitor). The data were expressed relative to that in the control (vehicle) group. Significance was evaluated by ANOVA (followed by Dunn's multiple comparisons) or unpaired t-test when the within-lot data were analyzed, or repeated measures ANOVA (followed by Dunn's multiple comparisons) or paired t-test when data from all 3 lots were analyzed (p < 0.05). Results and Discussion In general, a) PRH cocktails significantly induced transporter mRNA expression in the following order OAT2 ≈ NTCP ≈ OCT1 > OATP2B1 and repressed mRNA expression in the following order OATP1B3 > OATP1B1; b) these changes translated into significant induction of OAT2 (T1-T3) and NTCP (T2-T3, in only two lots) activity at the 1× PRH concentration. Compared with the influx transporters, the induction of mRNA expression of efflux transporters was modest, with mRNA expression of MRP2 and BSEP being induced the most. Conclusion Once these data are verified through in vivo probe drug PK studies in pregnancy, they can be populated into physiologically based pharmacokinetic (PBPK) models to predict, for all trimesters of pregnancy, transporter-mediated clearance of any drug that is a substrate of the affected transporters.
Collapse
Affiliation(s)
| | | | - Jashvant D. Unadkat
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, United States
| |
Collapse
|
5
|
Chupp DP, Rivera CE, Zhou Y, Xu Y, Ramsey PS, Xu Z, Zan H, Casali P. A humanized mouse that mounts mature class-switched, hypermutated and neutralizing antibody responses. Nat Immunol 2024; 25:1489-1506. [PMID: 38918608 PMCID: PMC11291283 DOI: 10.1038/s41590-024-01880-3] [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: 11/20/2023] [Accepted: 05/18/2024] [Indexed: 06/27/2024]
Abstract
Humanized mice are limited in terms of modeling human immunity, particularly with regards to antibody responses. Here we constructed a humanized (THX) mouse by grafting non-γ-irradiated, genetically myeloablated KitW-41J mutant immunodeficient pups with human cord blood CD34+ cells, followed by 17β-estradiol conditioning to promote immune cell differentiation. THX mice reconstitute a human lymphoid and myeloid immune system, including marginal zone B cells, germinal center B cells, follicular helper T cells and neutrophils, and develop well-formed lymph nodes and intestinal lymphoid tissue, including Peyer's patches, and human thymic epithelial cells. These mice have diverse human B cell and T cell antigen receptor repertoires and can mount mature T cell-dependent and T cell-independent antibody responses, entailing somatic hypermutation, class-switch recombination, and plasma cell and memory B cell differentiation. Upon flagellin or a Pfizer-BioNTech coronavirus disease 2019 (COVID-19) mRNA vaccination, THX mice mount neutralizing antibody responses to Salmonella or severe acute respiratory syndrome coronavirus 2 Spike S1 receptor-binding domain, with blood incretion of human cytokines, including APRIL, BAFF, TGF-β, IL-4 and IFN-γ, all at physiological levels. These mice can also develop lupus autoimmunity after pristane injection. By leveraging estrogen activity to support human immune cell differentiation and maturation of antibody responses, THX mice provide a platform to study the human immune system and to develop human vaccines and therapeutics.
Collapse
Affiliation(s)
- Daniel P Chupp
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Invivyd, Waltham, MA, USA
| | - Carlos E Rivera
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yulai Zhou
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Yijiang Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Patrick S Ramsey
- Department of Obstetrics & Gynecology, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Zhenming Xu
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
| | - Hong Zan
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA
- Prellis Biologics, Berkeley, CA, USA
| | - Paolo Casali
- The Antibody Laboratory, Department of Microbiology, Immunology & Molecular Genetics, The University of Texas Long School of Medicine, San Antonio, TX, USA.
- Department of Medicine, The University of Texas Long School of Medicine, San Antonio, TX, USA.
| |
Collapse
|
6
|
Hua K, Liu D, Xu Q, Peng Y, Sun Y, He R, Luo R, Jin H. The role of hormones in the regulation of lactogenic immunity in porcine and bovine species. Domest Anim Endocrinol 2024; 88:106851. [PMID: 38733944 DOI: 10.1016/j.domaniend.2024.106851] [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: 10/20/2023] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Colostrum and milk offer a complete diet and vital immune protection for newborn mammals with developing immune systems. High immunoglobulin levels in colostrum serve as the primary antibody source for newborn piglets and calves. Subsequent milk feeding support continued local antibody protection against enteric pathogens, as well as maturation of the developing immune system and provide nutrients for newborn growth. Mammals have evolved hormonal strategies that modulate the levels of immunoglobulins in colostrum and milk to facilitate effective lactational immunity. In addition, hormones regulate the gut-mammary gland-secretory immunoglobulin A (sIgA) axis in pregnant mammals, controlling the levels of sIgA in milk, which serves as the primary source of IgA for piglets and helps them resist pathogens such as PEDV and TGEV. In the present study, we review the existing studies on the interactions between hormones and the gut-mammary-sIgA axis/lactogenic immunity in mammals and explore the potential mechanisms of hormonal regulation that have not been studied in detail, to draw attention to the role of hormones in influencing the immune response of pregnant and lactating mammals and their offspring, and highlight the effect of hormones in regulating sIgA-mediated anti-infection processes in colostrum and milk. Discussion of the relationship between hormones and lactogenic immunity may lead to a better way of improving lactogenic immunity by determining a better injection time and developing new vaccines.
Collapse
Affiliation(s)
- Kexin Hua
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Dan Liu
- China Institute of Veterinary Drug Control, Beijing 100081, PR China
| | - Qianshuai Xu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Yuna Peng
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Yu Sun
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Rongrong He
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Rui Luo
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China
| | - Hui Jin
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, 430070, PR China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, PR China.
| |
Collapse
|
7
|
Feng S, Dai B, Li H, Fu H, Zhou Y. Efficacy of cognitive behavioral therapy for insomnia in the perinatal period: a meta-analysis of randomized controlled trials. Sleep Biol Rhythms 2024; 22:207-215. [PMID: 38524162 PMCID: PMC10959878 DOI: 10.1007/s41105-023-00502-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 11/12/2023] [Indexed: 03/26/2024]
Abstract
The meta-analysis aims to explore the effect of cognitive behavioral therapy for insomnia (CBT-I) in the perinatal period. Randomized controlled trials (RCTs) assessed the effects of CBT-I in perinatal women with insomnia, published in English, were eligible. Electronic searches were performed using PubMed, Embase (Elsevier), PsycINFO (Ebsco), and Web of Science (Clarivate Analytics). Insomnia Severity Index (ISI) as the primary outcome was used to estimate the pooled effects and durable efficacy of CBT-I. The secondary outcome measures were Edinburgh Postnatal Depression Scale (EPDS) and Pittsburgh Sleep Quality Index (PSQI). Of 46 studies reviewed, seven studies met the inclusion criteria. The meta-analysis indicated significant improvement in insomnia as measured with the ISI (standardized mean difference (SMD) = - 0.62, 95% confidence intervals (CI) - 0.77, - 0.47, I2 = 28%). At the follow-up time point, the meta-analysis indicated the durable efficacy of CBT-I (SMD = - 0.47, 95% CI - 0.90, - 0.03, I2 = 73%). Definite improvement of CBT-I on EPDS (SMD = -0.31, 95% CI - 0.55, - 0.06, I2 = 33%) and PSQI (SMD = - 0.82, 95% CI - 1.27, - 0.38, I2 = 68%) score change post-intervention were found. In sub-analyses, CBT-I had similar effect sizes, independent of possible modifiers (study population, comparison group, delivery format, etc.). This meta-analysis demonstrates that CBT-I is effective in alleviating insomnia, depression, and sleep quality among perinatal women. It is equally important to find that CBT-I has a durable efficacy on insomnia in the perinatal period. However, it is necessary to include larger samples and conduct rigorous RCTs to further explore this issue. Supplementary Information The online version contains supplementary material available at 10.1007/s41105-023-00502-z.
Collapse
Affiliation(s)
- Shuya Feng
- School of Nursing, Qingdao University, Qingdao, Shandong China
| | - Bingqin Dai
- Shandong Provincial Center for Disease Control and Prevention, Jinan, Shandong China
| | - Huawei Li
- School of Nursing, Qingdao University, Qingdao, Shandong China
| | - Huili Fu
- School of Nursing, Qingdao University, Qingdao, Shandong China
| | - Yunping Zhou
- School of Nursing, Qingdao University, Qingdao, Shandong China
| |
Collapse
|
8
|
Authement AK, Isoherranen N. The impact of pregnancy and associated hormones on the pharmacokinetics of Δ 9-tetrahydrocannabinol. Expert Opin Drug Metab Toxicol 2024; 20:73-93. [PMID: 38258511 DOI: 10.1080/17425255.2024.2309213] [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: 10/24/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
INTRODUCTION (-)-Δ9-tetrahydrocannabinol (THC) is the main psychoactive component of cannabis. Cannabis is the most widely used drug of abuse by pregnant individuals, but its maternal-fetal safety is still unclear. The changes in THC disposition during pregnancy may affect THC safety and pharmacology. AREAS COVERED This review summarizes the current literature on THC metabolism and pharmacokinetics in humans. It provides an analysis of how hormonal changes during pregnancy may alter the expression of cannabinoid metabolizing enzymes and THC and its metabolite pharmacokinetics. THC is predominately (>70%) cleared by hepatic metabolism to its psychoactive active metabolite, 11-OH-THC by cytochrome P450 (CYP) 2C9 and to other metabolites (<30%) by CYP3A4. Other physiological processes that change during pregnancy and may alter cannabinoid disposition are also reviewed. EXPERT OPINION THC and its metabolites disposition likely change during pregnancy. Hepatic CYP2C9 and CYP3A4 are induced in pregnant individuals and in vitro by pregnancy hormones. This induction of CYP2C9 and CYP3A4 is predicted to lead to altered THC and 11-OH-THC disposition and pharmacodynamic effects. More in vitro studies of THC metabolism and induction of the enzymes metabolizing cannabinoids are necessary to improve the prediction of THC pharmacokinetics in pregnant individuals.
Collapse
Affiliation(s)
- Aurora K Authement
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
9
|
Abdulrazaq AA, Ainsworth AJ, Britton JW, Shenoy CC, Babayev SN, Cascino GD, Smith KM. Seizure control in women with epilepsy undergoing assisted reproductive technology. Epilepsia 2023; 64:e207-e213. [PMID: 37596834 DOI: 10.1111/epi.17748] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
The objective of this study was to determine seizure control in women with epilepsy (WWE) undergoing assisted reproductive technology (ART). Through retrospective chart review, WWE undergoing ART were identified. Demographics and details regarding epilepsy type, seizure control, and ART procedures were extracted. Seizure frequency prior to and during ART were compared. We identified 12 WWE, who underwent 29 embryo transfers, resulting in 16 pregnancies and 10 live births. Nine women were seizure-free at least 2 years before fertility treatment, including three with resolved epilepsy. Seven were on antiseizure medications throughout fertility treatment and pregnancy, with only one on polytherapy. Eleven (all with controlled epilepsy or epilepsy in remission) remained seizure-free throughout fertility treatment. One woman with drug-resistant epilepsy continued to have seizures throughout fertility treatment and pregnancy without an exacerbation of seizure frequency. There was no increased seizure frequency associated with fertility treatment and subsequent pregnancy in this small series of WWE. Although this study was statistically underpowered, our results provide some preliminary evidence that ART might not pose a threat to seizure control, but larger, confirmatory studies are necessary.
Collapse
Affiliation(s)
| | | | | | - Chandra C Shenoy
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir N Babayev
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kelsey M Smith
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
10
|
Servin-Barthet C, Martínez-García M, Pretus C, Paternina-Die M, Soler A, Khymenets O, Pozo ÓJ, Leuner B, Vilarroya O, Carmona S. The transition to motherhood: linking hormones, brain and behaviour. Nat Rev Neurosci 2023; 24:605-619. [PMID: 37612425 DOI: 10.1038/s41583-023-00733-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2023] [Indexed: 08/25/2023]
Abstract
We are witnessing a stark increase in scientific interest in the neurobiological processes associated with pregnancy and maternity. Convergent evidence suggests that around the time of labour, first-time mothers experience a specific pattern of neuroanatomical changes that are associated with maternal behaviour. Here we provide an overview of the human neurobiological adaptations of motherhood, focusing on the interplay between pregnancy-related steroid and peptide hormones, and neuroplasticity in the brain. We discuss which brain plasticity mechanisms might underlie the structural changes detected by MRI, which hormonal systems are likely to contribute to such neuroanatomical changes and how these brain mechanisms may be linked to maternal behaviour. This Review offers an overarching framework that can serve as a roadmap for future investigations.
Collapse
Affiliation(s)
- Camila Servin-Barthet
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
| | - Magdalena Martínez-García
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Clara Pretus
- Hospital del Mar Research Institute, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de els Ciències de la Salut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Paternina-Die
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Soler
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain
- Hospital del Mar Research Institute, Barcelona, Spain
| | | | - Óscar J Pozo
- Hospital del Mar Research Institute, Barcelona, Spain
| | - Benedetta Leuner
- Psychology Department, The Ohio State University, Columbus, OH, USA
| | - Oscar Vilarroya
- Unitat de Recerca en Neurociència Cognitiva, Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona, Barcelona, Spain.
- Hospital del Mar Research Institute, Barcelona, Spain.
| | - Susana Carmona
- Instituto de Investigación Sanitaria Gregorio Marañon, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
11
|
Giri T, Panda S, Palanisamy A. Pregnancy-induced differential expression of SARS-CoV-2 and influenza a viral entry factors in the lower respiratory tract. PLoS One 2023; 18:e0281033. [PMID: 37437040 DOI: 10.1371/journal.pone.0281033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Despite differences in the clinical presentation of coronavirus disease-19 and pandemic influenza in pregnancy, fundamental mechanistic insights are currently lacking because of the difficulty in recruiting critically ill pregnant subjects for research studies. Therefore, to better understand host-pathogen interaction during pregnancy, we performed a series of foundational experiments in pregnant rats at term gestation to assess the expression of host entry factors for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza A virus (IAV) and genes associated with innate immune response in the lower respiratory tract. We report that pregnancy is characterized by a decrease in host factors mediating SARS-CoV-2 entry and an increase in host factors mediating IAV entry. Furthermore, using flow cytometric assessment of immune cell populations and immune provocation studies, we show an increased prevalence of plasmacytoid dendritic cells and a Type I interferon-biased environment in the lower respiratory tract of pregnancy, contrary to the expected immunological indolence. Our findings, therefore, suggest that the dissimilar clinical presentation of COVID-19 and pandemic influenza A in pregnancy could partly be due to differences in the extent of innate immune activation from altered viral tropism and indicate the need for comparative mechanistic investigations with live virus studies.
Collapse
Affiliation(s)
- Tusar Giri
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Santosh Panda
- Department of Pathology, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Arvind Palanisamy
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States of America
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, United States of America
| |
Collapse
|
12
|
Dukic J, Ehlert U. Longitudinal Course of Sex Steroids From Pregnancy to Postpartum. Endocrinology 2023; 164:bqad108. [PMID: 37450580 PMCID: PMC10499333 DOI: 10.1210/endocr/bqad108] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
CONTEXT Sex steroids (SS) typically rise during pregnancy and decline after birth, but no consistent reference values exist for these hormonal courses. We aimed to establish an overview of SS secretion patterns during the peripartum and to better understand how SS contribute to maternal and fetal pathologies. EVIDENCE ACQUISITION A systematic literature search was conducted in accordance with the PRISMA guidelines using PubMed, Cochrane Library, and PsycINFO. Additionally, we conducted a supplementary manual search of references. Observational studies published in English and assessing estradiol, progesterone, and testosterone over the course of the peripartum in physically healthy female subjects were included, without restrictions on year of publication. Extracted data were analyzed descriptively and visually. EVIDENCE SYNTHESIS SS increase progressively during pregnancy, with an extremely wide range of reported concentrations, especially in the third trimester. In fact, reported concentrations varied up to 5000-fold at comparable measurement time points. CONCLUSIONS A comprehensive understanding of the influence of SS levels on associated maternal and fetal pathologies is currently hindered by 2 main factors. First, reported SS levels vary widely during the peripartum period. Second, the current state of knowledge on how SS are associated with pathologies in mothers and babies is largely based on correlational studies, and causality thus remains unclear. Consequently, we recommend the development of a systematic reference framework that follows the suggestions presented in this review. This would enable the establishment of SS reference values for a healthy population, resulting in the possibility to draw conclusions about deviations and related pathologies.
Collapse
Affiliation(s)
- Jelena Dukic
- Department of Clinical Psychology and Psychotherapy, University of Zurich, 8050 Zurich, Switzerland
| | - Ulrike Ehlert
- Department of Clinical Psychology and Psychotherapy, University of Zurich, 8050 Zurich, Switzerland
| |
Collapse
|
13
|
Barrett JS, Azer K. Opportunities for Systems Biology and Quantitative Systems Pharmacology to Address Knowledge Gaps for Drug Development in Pregnancy. J Clin Pharmacol 2023; 63 Suppl 1:S96-S105. [PMID: 37317502 DOI: 10.1002/jcph.2265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 04/25/2023] [Indexed: 06/16/2023]
Abstract
Pregnant women are still viewed as therapeutic orphans to the extent that they are avoided as participants in mainstream clinical trials and not considered a priority for targeted drug research despite the fact that many clinical conditions exist during pregnancy for which pharmacotherapy is warranted. Part of the challenge is the uncertain risk potential that pregnant women represent in the absence of timely and costly toxicology and developmental pharmacology studies, which only partly mitigate such risks. Even when clinical trials are conducted in pregnant women, they are often underpowered and absent biomarkers and exclude evaluation across multiple stages of pregnancy where relevant development risk could have been assessed. Quantitative systems pharmacology model development has been proposed as one solution to fill knowledge gaps, make earlier and perhaps more informed risk assessment, and design more informative trials with better recommendations for biomarker and end point selection including design and sample size optimality. Funding for translational research in pregnancy is limited but will fill some of these gaps, especially when joined with ongoing clinical trials in pregnancy that also fill certain knowledge gaps, especially biomarker and end point evaluation across pregnancy states linked to clinical outcomes. Opportunities exist for further advances in quantitative systems pharmacology model development with the inclusion of real-world data sources and complimentary artificial intelligence/machine learning approaches. The successful coordination of the approach reliant on these new data sources will require commitments to share data and a diverse multidisciplinary group that seeks to develop open science models that benefit the entire research community, ensuring that such models can be used with high fidelity. New data opportunities and computational resources are highlighted in an effort to project how these efforts can move forward.
Collapse
Affiliation(s)
| | - Karim Azer
- Axcella Therapeutics, Cambridge, Massachusetts, USA
| |
Collapse
|
14
|
Norris JN, Waack AL, Becker KN, Keener M, Hoyt A, Reinard K. Glioblastoma in pregnant patient with pathologic and exogenous sex hormone exposure and family history of high-grade glioma: A case report and review of the literature. Surg Neurol Int 2023; 14:169. [PMID: 37292394 PMCID: PMC10246315 DOI: 10.25259/sni_58_2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Abstract
Background Glioblastoma (GBM) incidence is higher in males, suggesting sex hormones may influence GBM tumorigenesis. Patients with GBM and altered sex hormone states could offer insight into a relationship between the two. Most GBMs arise sporadically and heritable genetic influence on GBM development is poorly understood, but reports describing familial GBM suggest genetic predispositions exist. However, no existing reports examine GBM development in context of both supraphysiologic sex hormone states and familial predisposition for GBM. We present a case of isocitrate dehydrogenase (IDH)-wild type GBM in a young pregnant female with polycystic ovary syndrome (PCOS), history of in vitro fertilization (IVF), and significant family history of GBM and further discuss how unique sex hormone states and genetics may affect GBM development or progression. Case Description A 35-year-old pregnant female with PCOS and recent history of IVF treatment and frozen embryo transfer presented with seizure and headache. Imaging revealed a right frontal brain mass. Molecular and histopathological analysis of the resected tumor supported a diagnosis of IDH-wild type GBM. The patient's family medical history was significant for GBM. Current literature indicates testosterone promotes GBM cell proliferation, while estrogen and progesterone effects vary with receptor subtype and hormone concentration, respectively. Conclusion Sex hormones and genetics likely exert influence on GBM development and progression that may compound with concurrence. Here, we describe a unique case of GBM in a young pregnant patient with a family history of glioma and atypical sex hormone exposure due to endocrine disorder and pregnancy assisted by exogenous IVF hormone administration.
Collapse
Affiliation(s)
- Jordan N. Norris
- Department of Surgery, Division of Neurosurgery, University of Toledo, Ohio, United States
| | - Andrew L. Waack
- Department of Surgery, Division of Neurosurgery, University of Toledo, Ohio, United States
| | - Kathryn N. Becker
- Department of Surgery, Division of Neurosurgery, University of Toledo, Ohio, United States
| | - Myles Keener
- Department of Surgery, Division of Neurosurgery, University of Toledo, Ohio, United States
| | - Alastair Hoyt
- Department of Surgery, Division of Neurosurgery, University of Toledo, Ohio, United States
| | - Kevin Reinard
- Department of Neurosurgery, Promedica, Toledo, Ohio, United States
| |
Collapse
|
15
|
Levin G, Ein-Dor T. A unified model of the biology of peripartum depression. Transl Psychiatry 2023; 13:138. [PMID: 37117197 PMCID: PMC10147643 DOI: 10.1038/s41398-023-02439-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023] Open
Abstract
Peripartum depression (PPD) is a prevalent and debilitating disorder that adversely affects the development of mothers and infants. Recently, there has been a plea for increased mental health screening during the peripartum period; however, currently, there is no accurate screening tool to identify women at risk of PPD. In addition, some women do not respond to current treatment schemes and develop treatment-resistant depression. The current perspective aims to propose a unified understanding of the biological underpinnings of PPD (UmPPD) that considers the heterogeneity in the onset, symptoms cluster, and severity of PPD. Such a model could promote basic and applied research on PPD and suggest new treatment avenues. The central hub of the model is the kynurenine pathway (KP) and the KP-serotonin ratio. The forces and specific processes at play that cause an imbalance within the KP and between KP and serotonin are inflammation, stress, reproductive hormones (especially estradiol and progesterone), and oxytocin. UmPPD predicts that the most severe PPD would comprise prolonged inflammation, ongoing or multiple stressors, excessive estrogen, progesterone resistance, and avoidance of breastfeeding, skin-to-skin contact, and social proximity. These factors would be associated with a higher likelihood of developing PPD, early onset, and more significant symptom severity. In addition, subtypes of PPD would consist of different compositions and expressions of these components, with one central common factor. UmPPD could aid in directing future research and possibly detecting critical processes that could help discover, develop, and utilize novel treatments for PPD.
Collapse
Affiliation(s)
- Gal Levin
- Reichman University. Please address all correspondence to: Tsachi Ein-Dor, Baruch Ivcher School of Psychology, Reichman University, University St. 8, Herzliya, 4610101, Israel
| | - Tsachi Ein-Dor
- Reichman University. Please address all correspondence to: Tsachi Ein-Dor, Baruch Ivcher School of Psychology, Reichman University, University St. 8, Herzliya, 4610101, Israel.
| |
Collapse
|
16
|
Utami AM, Horbach SE, Meijer-Jorna LB, Waas IS, de Boer OJ, van der Wal AC, van der Horst CM. Microvascular proliferation in arteriovenous malformation of the hand worsens during pregnancy: a case report. Ann Med Surg (Lond) 2023; 85:1262-1269. [PMID: 37113922 PMCID: PMC10129217 DOI: 10.1097/ms9.0000000000000507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/18/2023] [Indexed: 04/29/2023] Open
Abstract
Arteriovenous malformations (AVMs) are rare congenital disorders characterized by episodes of disproportionate growth that can cause pain and severe bleeding, with microvascular proliferation (MVP) associated with these episodes. Hormonal influences can also worsen the symptoms in patients with AVM. Case presentation This case report presents a female patient with congenital vascular malformations of the left hand since birth, whose symptoms worsened during puberty and pregnancy, ultimately leading to amputation of the left hand due to unbearable pain and loss of function. Pathologic analysis revealed substantial MVP activity within the tissues of the AVM, with an expression of receptors for estrogen, growth hormone, and follicle-stimulating hormone in the vessels of the AVM, including MVP areas. Resected materials not related to pregnancy revealed chronic inflammation and fibrosis but hardly any MVP. Discussion and conclusion These findings suggest a role for MVP in the progressive growth of AVM during pregnancy, with a potential role for hormonal influences. The case highlights the relationship between AVM symptoms and size during pregnancy and the pathological findings of MVP areas within the AVM with hormone receptor expression on proliferating vessels in resected materials.
Collapse
Affiliation(s)
- Amalia M. Utami
- Department of Pathology
- Department of Pathology Anatomy, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
- Corresponding author. Address: Department of Pathology Anatomy, Faculty of Medicine, Hasanuddin University, Jln. Perintis Kemerdekaan Km.10, Makassar 90245, Indonesia. Tel./fax: +62411-586010. E-mail address: (A.M. Utami)
| | - Sophie E.R. Horbach
- Department of Plastic Surgery, Amsterdam University Medical Center, AMC, University of Amsterdam, Amsterdam
| | | | | | | | | | | |
Collapse
|
17
|
Puglisi S, Basile V, Sperone P, Terzolo M. Pregnancy in patients with adrenocortical carcinoma: a case-based discussion. Rev Endocr Metab Disord 2023; 24:85-96. [PMID: 36414840 DOI: 10.1007/s11154-022-09769-y] [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] [Accepted: 10/24/2022] [Indexed: 11/24/2022]
Abstract
Although adrenocortical carcinoma (ACC) during pregnancy is rare, a retrospective review of a case series at our hospital revealed that almost one third of our patients were women in childbearing age. Given that the age of maternity is increasing, dealing with a tumor diagnosis during pregnancy and the need for fertility planning in cancer survivors is likely to become more frequent.We thus carried out a case-based discussion regarding: i) diagnosing and treating an ACC during pregnancy; ii) patients conceiving while on mitotane; iii) ACC survivors with a maternal desire.In each of these cases, it is important to provide patients with sufficient information, to offer medical advice and psychological support, to personalize treatments in accordance with the wishes of the patient and her relatives, and to collaborate with other specialists since a multidisciplinary expert team is required to manage each case individually.
Collapse
Affiliation(s)
- Soraya Puglisi
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Vittoria Basile
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy
| | - Paola Sperone
- Medical Oncology, Department of Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Italy
| | - Massimo Terzolo
- Internal Medicine, Department of Clinical and Biological Sciences, San Luigi Gonzaga Hospital, University of Turin, Regione Gonzole 10, 10043, Orbassano, Italy.
| |
Collapse
|
18
|
Soedirdjo SDH, Rodriguez LA, Chung YC, Casey E, Dhaher YY. Sex hormone-mediated change on muscle activation deactivation dynamics in young eumenorrheic women. Front Physiol 2023; 14:1104578. [PMID: 36960149 PMCID: PMC10029997 DOI: 10.3389/fphys.2023.1104578] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/22/2023] [Indexed: 03/09/2023] Open
Abstract
The goal of the study was to characterize muscle activation/deactivation dynamics across the menstrual cycle in healthy young women. Twenty-two healthy eumenorrheic women (age: 27.0 ± 4.4 years; mean ± SD) were tested every other day for one menstrual cycle. Serum estradiol and progesterone were quantified at the time of testing. Peak torque (PT), time to peak torque (TPT), and half relaxation time (HRT) of soleus muscle twitch were measured. Muscle twitch was elicited by delivering 1 ms width electrical pulses to the tibial nerve at an intensity that generated a maximum motor response (S-100) and at supramaximal intensity (S-120; 1.2 × S-100). The analyses were performed for each menstrual cycle phase: 1) the follicular phase to analyze the effect of estradiol while the progesterone concentrations remained at low concentrations; 2) the luteal phase to analyze the effect of progesterone with background estradiol concentrations. In the follicular phase, there was no association of estradiol for PT, TPT, and HRT. In the luteal phase, while estradiol had no association on PT, TPT, and HRT, progesterone expressed a significant association with HRT reduction but no association on PT or TPT. Also, there was a significant estradiol and progesterone interaction for HRT. However, the regression parameters are nearly zero, suggesting that the change in HRT may not have an impact on muscle performance across the menstrual cycle but implications on other women's health conditions with elevated sex hormone concentrations, such as pregnancy, may prove critical.
Collapse
Affiliation(s)
- Subaryani D. H. Soedirdjo
- Department of Physical Medicine and Rehabilitation, UT Southwestern Medical Center, Dallas, TX, United States
| | - Luis A. Rodriguez
- Department of Physical Medicine and Rehabilitation, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
- Department of Orthopedic Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - Yu-Chen Chung
- Department of Physical Medicine and Rehabilitation, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ellen Casey
- Department of Physiatry, Hospital for Special Surgery, New York, NY, United States
| | - Yasin Y. Dhaher
- Department of Physical Medicine and Rehabilitation, UT Southwestern Medical Center, Dallas, TX, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
- Department of Orthopedic Surgery, UT Southwestern Medical Center, Dallas, TX, United States
- *Correspondence: Yasin Y. Dhaher,
| |
Collapse
|
19
|
Fronto-parietal alpha ERD and visuo-spatial attention in pregnant women. Brain Res 2022; 1798:148130. [DOI: 10.1016/j.brainres.2022.148130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/27/2022] [Accepted: 10/22/2022] [Indexed: 11/20/2022]
|
20
|
Fashe MM, Fallon JK, Miner TA, Tiley JB, Smith PC, Lee CR. Impact of pregnancy related hormones on drug metabolizing enzyme and transport protein concentrations in human hepatocytes. Front Pharmacol 2022; 13:1004010. [PMID: 36210832 PMCID: PMC9532936 DOI: 10.3389/fphar.2022.1004010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Pregnancy alters the disposition and exposure to multiple drugs indicated for pregnancy-related complications. Previous in vitro studies have shown that pregnancy-related hormones (PRHs) alter the expression and function of certain cytochrome P450s (CYPs) in human hepatocytes. However, the impact of PRHs on hepatic concentrations of non-CYP drug-metabolizing enzymes (DMEs) and transport proteins remain largely unknown. In this study, sandwich-cultured human hepatocytes (SCHH) from five female donors were exposed to vehicle or PRHs (estrone, estradiol, estriol, progesterone, cortisol, and placental growth hormone), administered individually or in combination, across a range of physiologically relevant PRH concentrations for 72 h. Absolute concentrations of 33 hepatic non-CYP DMEs and transport proteins were quantified in SCHH membrane fractions using a quantitative targeted absolute proteomics (QTAP) isotope dilution nanoLC-MS/MS method. The data revealed that PRHs altered the absolute protein concentration of various DMEs and transporters in a concentration-, isoform-, and hepatocyte donor-dependent manner. Overall, eight of 33 (24%) proteins exhibited a significant PRH-evoked net change in absolute protein concentration relative to vehicle control (ANOVA p < 0.05) across hepatocyte donors: 1/11 UGTs (9%; UGT1A4), 4/6 other DMEs (67%; CES1, CES2, FMO5, POR), and 3/16 transport proteins (19%; OAT2, OCT3, P-GP). An additional 8 (24%) proteins (UGT1A1, UGT2B4, UGT2B10, FMO3, OCT1, MRP2, MRP3, ENT1) exhibited significant PRH alterations in absolute protein concentration within at least two individual hepatocyte donors. In contrast, 17 (52%) proteins exhibited no discernable impact by PRHs either within or across hepatocyte donors. Collectively, these results provide the first comprehensive quantitative proteomic evaluation of PRH effects on non-CYP DMEs and transport proteins in SCHH and offer mechanistic insight into the altered disposition of drug substrates cleared by these pathways during pregnancy.
Collapse
Affiliation(s)
- Muluneh M. Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Taryn A. Miner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jacqueline B. Tiley
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
21
|
Expression of Key Steroidogenic Enzymes in Human Placenta and Associated Adverse Pregnancy Outcomes. MATERNAL-FETAL MEDICINE 2022. [DOI: 10.1097/fm9.0000000000000167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
22
|
Zhao W, Zhang J, Feng A, Yin H, Liu C, Pan Y. Rapid Quantification of Endogenous Steroids in Human Serum Using Leidenfrost Effect-Assisted Thermal Desorption Atmospheric Pressure Photoionization Orbitrap Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2022; 33:1250-1259. [PMID: 35748155 DOI: 10.1021/jasms.2c00077] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Unconjugated sex steroids in human serum play a crucial role in physiological and pathological studies and are frequently considered as biomarkers in clinical diagnosis. Because of their low polarity, poor volatility, and low concentration, the rapid and highly sensitive analysis of sex steroids in real serum matrix by ambient mass spectrometry is still challenging. Here, Leidenfrost effect-assisted thermal desorption atmospheric pressure photoionization orbitrap mass spectrometry (LETD-APPI-MS) was developed and applied to quantify free sex steroids in human serum without derivatization and chromatography separation within a few minutes. The concentration of target analyte could be increased by approximately two orders during the LETD process. The limit of quantifications and detections of endogenous sex steroids in human serum were measured at the ppt level. In contrast with commonly used immunoassays in clinical laboratories, LETD-APPI-MS enables the accurate measurements of multiple free sex steroids without the interference of cross-reactions. The endogenous sex steroids of 38 female serums at four physiological stages during pregnancy were rapidly tested by LETD-APPI-MS, whose results were highly consistent with that using liquid chromatography-atmospheric pressure chemical ionization mass spectrometry (LC-APCI-MS), indicating LETD-APPI-MS has a strong clinical application potential in steroid analysis.
Collapse
Affiliation(s)
- Wan Zhao
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei 230029, China
| | - Jing Zhang
- Anhui Prevention and Treatment Center for Occupational Disease, Anhui No. 2 Provincial People's Hospital, Hefei 230041, China
| | - Airong Feng
- Instruments center for physical science, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Hao Yin
- Instruments center for physical science, University of Science and Technology of China, Hefei, Anhui 230026, P.R. China
| | - Chengyuan Liu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei 230029, China
| | - Yang Pan
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei 230029, China
| |
Collapse
|
23
|
Cervantes O, Talavera IC, Every E, Coler B, Li M, Li A, Li H, Adams Waldorf K. Role of hormones in the pregnancy and sex-specific outcomes to infections with respiratory viruses. Immunol Rev 2022; 308:123-148. [PMID: 35373371 PMCID: PMC9189035 DOI: 10.1111/imr.13078] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 01/13/2023]
Abstract
Pregnant women infected with pathogenic respiratory viruses, such as influenza A viruses (IAV) and coronaviruses, are at higher risk for mortality, hospitalization, preterm birth, and stillbirth. Several factors are likely to contribute to the susceptibility of pregnant individuals to severe lung disease including changes in pulmonary physiology, immune defenses, and effector functions of some immune cells. Pregnancy is also a physiologic state characterized by higher levels of multiple hormones that may impact the effector functions of immune cells, such as progesterone, estrogen, human chorionic gonadotropin, prolactin, and relaxin. Each of these hormones acts to support a tolerogenic immune state of pregnancy, which helps prevent fetal rejection, but may also contribute to an impaired antiviral response. In this review, we address the unique role of adaptive and innate immune cells in the control of pathogenic respiratory viruses and how pregnancy and specific hormones can impact their effector actions. We highlight viruses with sex-specific differences in infection outcomes and why pregnancy hormones may contribute to fetal protection but aid the virus at the expense of the mother's health.
Collapse
Affiliation(s)
- Orlando Cervantes
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Irene Cruz Talavera
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, United States of America
| | - Emma Every
- University of Washington School of Medicine, Spokane, Washington, United States of America
| | - Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Elson S. Floyd College of Medicine, Washington State University, Spokane, Washington, United States of America
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Department of Biological Sciences, Columbia University, New York City, New York, United States of America
| | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
- Case Western Reserve, Cleveland, Ohio, United States of America
| | - Hanning Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| | - Kristina Adams Waldorf
- Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
24
|
Bushby EV, Cotter SC, Wilkinson A, Friel M, Collins LM. Judgment Bias During Gestation in Domestic Pigs. Front Vet Sci 2022; 9:881101. [PMID: 35647100 PMCID: PMC9133791 DOI: 10.3389/fvets.2022.881101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/12/2022] [Indexed: 11/21/2022] Open
Abstract
In humans and rats, changes in affect are known to occur during pregnancy, however it is unknown how gestation may influence mood in other non-human mammals. This study assessed changes in pigs' judgment bias as a measure of affective state throughout gestation. Pigs were trained to complete a spatial judgment bias task with reference to positive and negative locations. We tested gilts before mating, and during early and late gestation, by assessing their responses to ambiguous probe locations. Pigs responded increasingly negatively to ambiguous probes as gestation progressed and there were consistent inter-individual differences in baseline optimism. This suggests that the pigs' affective state may be altered during gestation, although as a non-pregnant control group was not tested, an effect of learning cannot be ruled out. These results suggest that judgment bias is altered during gestation in domestic pigs, consequently raising novel welfare considerations for captive multiparous species.
Collapse
Affiliation(s)
- Emily V. Bushby
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Sheena C. Cotter
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Anna Wilkinson
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
| | - Mary Friel
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Lisa M. Collins
- School of Life Sciences, University of Lincoln, Lincoln, United Kingdom
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- *Correspondence: Lisa M. Collins
| |
Collapse
|
25
|
Bai M, Chen M, Zeng Q, Lu S, Li P, Ma Z, Lin N, Zheng C, Zhou H, Zeng S, Sun D, Jiang H. Up‐regulation of hepatic CD36 by increased corticosterone/cortisol levels via GR leads to lipid accumulation in liver and hypertriglyceridaemia during pregnancy. Br J Pharmacol 2022; 179:4440-4456. [PMID: 35491243 DOI: 10.1111/bph.15863] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/06/2022] [Accepted: 04/21/2022] [Indexed: 11/29/2022] Open
Affiliation(s)
- Mengru Bai
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Mingyang Chen
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Qingquan Zeng
- Women's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Shuanghui Lu
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Ping Li
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Zhiyuan Ma
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Nengming Lin
- Department of Clinical Pharmacy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Caihong Zheng
- Women's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Hui Zhou
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Su Zeng
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| | - Dongli Sun
- Women's Hospital, School of Medicine Zhejiang University Hangzhou China
| | - Huidi Jiang
- Laboratory of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences Zhejiang University Hangzhou China
| |
Collapse
|
26
|
Le A, Huang KJ, Cirrincione LR. Regulation of drug-metabolizing enzymes by sex-related hormones: clinical implications for transgender medicine. Trends Pharmacol Sci 2022; 43:582-592. [PMID: 35487786 DOI: 10.1016/j.tips.2022.03.006] [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: 11/07/2021] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
Transgender medicine is a diverse and growing clinical field with unmet gaps in pharmacological knowledge. Hormone therapy (testosterone or estrogen treatment), one part of the standard of medical care for transgender adults, aligns secondary sex characteristics with an individual's gender identity and expression. Despite established effects of sex steroids on drug-metabolizing enzyme expression and activity in vitro and in animal models, the effect of long-term, supraphysiological sex hormone treatment on drug metabolism in transgender adults is not yet established. Here, we synthesize available in vitro and animal model data with pharmacological concepts in transgender medicine to predict potential effects of sex steroids on drug-metabolizing enzymes, and their relationship with potential hormone-drug interactions, in transgender medicine.
Collapse
Affiliation(s)
- An Le
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kai J Huang
- Center for Transyouth Health and Development, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | | |
Collapse
|
27
|
O’Brien LM. Sleep in Pregnancy. Respir Med 2022. [DOI: 10.1007/978-3-030-93739-3_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Yu B, Guo F, Yang Y, Long W, Zhou J. Steroidomics of Pregnant Women at Advanced Age. Front Endocrinol (Lausanne) 2022; 13:796909. [PMID: 35282454 PMCID: PMC8905515 DOI: 10.3389/fendo.2022.796909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 02/01/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To discover the profiles of different steroid hormones at the maternal-fetal interface and reveal the change characteristics in pregnant women at advanced maternal age (AMA). METHODS Forty pregnant women were recruited in the study, including 20 AMA women (age ≥ 35) and 20 normal controls (age < 35 and without pregnancy complications). Among AMA women, 6 (AMA2) had pregnancy complications, and 14 (AMA1) had no complications. Their maternal blood (MB), placental tissue (P), and fetal cord blood (CB) were collected, and 18 different steroid hormone metabolites were analyzed by liquid chromatography tandem mass spectrometry (LC-MS/MS). RESULTS The estradiol (E2) levels in MB were higher than those in P and CB. In contrast, the estrone (E1) and estriol (E3) levels were higher in P and CB. Compared with the progesterone levels (P4) in MB, those in P and CB were higher; however, cortisol (F) levels were deficient. In contrast, F in MB was maintained at an elevated level. Further, cortisone (E) levels in CB were higher than those in MB and P. Except for the decline of testosterone (T), androstenedione (A2) and Dihydrotestosterone (DHT), there were no significant differences in the other 15 steroid hormones in MB between the AMA1 and the control group (p>0.05). Compared with the AMA1 group, androgen levels were significantly higher in AMA2, especially in T (1.55 vs. 0.68 ng/ml, p=0.023), A2 (2.27 vs. 0.92 ng/ml, p=0.011) and Dehydroepiandrosterone (DHEA) (2.39 vs. 1.50 ng/ml, p=0.028). However, there were no significant changes in P and CB between two groups. CONCLUSION There are distribution rules and cascade changes of steroid profiles in maternal-fetal compartments. Significantly high androgen levels in AMA women have a positive relationship with adverse pregnancy complications.
Collapse
Affiliation(s)
- Bin Yu
- *Correspondence: Bin Yu, ; Jun Zhou,
| | | | | | | | - Jun Zhou
- *Correspondence: Bin Yu, ; Jun Zhou,
| |
Collapse
|
29
|
The Neuroactive Steroid Pregnanolone Glutamate: Anticonvulsant Effect, Metabolites and Its Effect on Neurosteroid Levels in Developing Rat Brains. Pharmaceuticals (Basel) 2021; 15:ph15010049. [PMID: 35056106 PMCID: PMC8780580 DOI: 10.3390/ph15010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/02/2022] Open
Abstract
Pregnanolone glutamate (PA-G) is a neuroactive steroid that has been previously demonstrated to be a potent neuroprotective compound in several biological models in vivo. Our in vitro experiments identified PA-G as an inhibitor of N-methyl-D-aspartate receptors and a potentiator of γ-aminobutyric acid receptors (GABAARs). In this study, we addressed the hypothesis that combined GABAAR potentiation and NMDAR antagonism could afford a potent anticonvulsant effect. Our results demonstrated the strong age-related anticonvulsive effect of PA-G in a model of pentylenetetrazol-induced seizures. PA-G significantly decreased seizure severity in 12-day-old animals, but only after the highest dose in 25-day-old animals. Interestingly, the anticonvulsant effect of PA-G differed both qualitatively and quantitatively from that of zuranolone, an investigational neurosteroid acting as a potent positive allosteric modulator of GABAARs. Next, we identified 17-hydroxy-pregnanolone (17-OH-PA) as a major metabolite of PA-G in 12-day-old animals. Finally, the administration of PA-G demonstrated direct modulation of unexpected neurosteroid levels, namely pregnenolone and dehydroepiandrosterone sulfate. These results suggest that compound PA-G might be a pro-drug of 17-OH-PA, a neurosteroid with a promising neuroprotective effect with an unknown mechanism of action that may represent an attractive target for studying perinatal neural diseases.
Collapse
|
30
|
Sundararajan A, Vora K, Saiyed S, Natesan S. Comparative profiling of prenatal cortisol and DHEA-S among pregnant women with poor birth outcome and pregnant women with normal birth outcome. Clin Endocrinol (Oxf) 2021; 95:863-872. [PMID: 34370329 DOI: 10.1111/cen.14569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 07/12/2021] [Accepted: 07/19/2021] [Indexed: 12/26/2022]
Abstract
CONTEXT Cortisol and dehydroepiandrosterone-sulfate (DHEA-S) are indispensable hormones for normal pregnancy. It is unclear if these hormones, specifically DHEA-S can offer value for predicting poor birth outcome. OBJECTIVE To compare prenatal cortisol and DHEA-S levels among pregnant women with normal or poor birth outcome. METHODS Plasma and saliva were collected prospectively from women in second-third trimester of pregnancy. Women with normal birth outcome (NBO) (n = 501) included live birth, no pregnancy complications and ≥2.5 kg infant birth weight. Women with poor birth outcome included adverse birth outcome (ABO) (n = 50) or low birth weight outcome (LBW) (n = 147). Enzyme-linked immunosorbent assay was performed to measure hormone concentrations in plasma and saliva. RESULTS Circulatory-DHEA-S levels in pregnant women with ABO were higher than women with NBO (p = .043). Among ABO, only stillbirth cases demonstrated significant increase in circulatory-DHEA-S levels (p = .006). Circulatory and salivary cortisol/DHEA-S ratio was lower among women with stillbirth (p = .004) and ABO outcome (p = .043) respectively compared with women with NBO. Consistently, increased odds of ABO were observed in pregnant women with highest circulatory-DHEA-S levels (odds ratio quartile score 1 vs. 4, 2.79, p = .027) and lowest salivary cortisol/DHEA-S ratio (score 4 vs. 2, 2.83, p = .025). Increased odds of stillbirth outcome were observed in pregnant women with highest circulatory-DHEA-S levels (odds ratio quartile score 1 vs. 4, 8.47, p = .046) and lowest circulatory cortisol/DHEA-S ratio (score 4 vs. 1, 4.803, p = .048). Associations remained significant after adjusting for confounders. Women with LBW did not demonstrate significant changes in cortisol or DHEA-S levels. CONCLUSION Prenatal measurement of DHEA-S or cortisol/DHEA-S ratio may offer significant value for predicting adverse birth, specifically stillbirth outcome.
Collapse
Affiliation(s)
| | - Kranti Vora
- Indian Institute of Public Health Gandhinagar, Lekawada, Gandhinagar, India
- University of Canberra, Canberra, Australian Capital Territory, Australia
| | - Shahin Saiyed
- Indian Institute of Public Health Gandhinagar, Lekawada, Gandhinagar, India
| | | |
Collapse
|
31
|
van Hoogdalem MW, Wexelblatt SL, Akinbi HT, Vinks AA, Mizuno T. A review of pregnancy-induced changes in opioid pharmacokinetics, placental transfer, and fetal exposure: Towards fetomaternal physiologically-based pharmacokinetic modeling to improve the treatment of neonatal opioid withdrawal syndrome. Pharmacol Ther 2021; 234:108045. [PMID: 34813863 DOI: 10.1016/j.pharmthera.2021.108045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 02/07/2023]
Abstract
Physiologically-based pharmacokinetic (PBPK) modeling has emerged as a useful tool to study pharmacokinetics (PK) in special populations, such as pregnant women, fetuses, and newborns, where practical hurdles severely limit the study of drug behavior. PK in pregnant women is variable and everchanging, differing greatly from that in their nonpregnant female and male counterparts typically enrolled in clinical trials. PBPK models can accommodate pregnancy-induced physiological and metabolic changes, thereby providing mechanistic insights into maternal drug disposition and fetal exposure. Fueled by the soaring opioid epidemic in the United States, opioid use during pregnancy continues to rise, leading to an increased incidence of neonatal opioid withdrawal syndrome (NOWS). The severity of NOWS is influenced by a complex interplay of extrinsic and intrinsic factors, and varies substantially between newborns, but the extent of prenatal opioid exposure is likely the primary driver. Fetomaternal PBPK modeling is an attractive approach to predict in utero opioid exposure. To facilitate the development of fetomaternal PBPK models of opioids, this review provides a detailed overview of pregnancy-induced changes affecting the PK of commonly used opioids during gestation. Moreover, the placental transfer of these opioids is described, along with their disposition in the fetus. Lastly, the implementation of these factors into PBPK models is discussed. Fetomaternal PBPK modeling of opioids is expected to provide improved insights in fetal opioid exposure, which allows for prediction of postnatal NOWS severity, thereby opening the way for precision postnatal treatment of these vulnerable infants.
Collapse
Affiliation(s)
- Matthijs W van Hoogdalem
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Scott L Wexelblatt
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Henry T Akinbi
- Perinatal Institute, Division of Neonatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Tomoyuki Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH, USA; Center for Addiction Research, College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
32
|
Guven B, Akdemir Y. The change pattern in serum G protein-coupled estrogen receptor-1 (GPER1) levels during pregnancy with and without gestational diabetes mellitus. Horm Mol Biol Clin Investig 2021; 43:207-210. [PMID: 34787384 DOI: 10.1515/hmbci-2021-0023] [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: 03/10/2021] [Accepted: 11/03/2021] [Indexed: 11/15/2022]
Abstract
OBJECTIVES The purpose of this study was to evaluate serum G protein-coupled estrogen receptor-1 (GPER1) levels in non-pregnant and pregnant with and without gestational diabetes mellitus (GDM). METHODS The study comprised 40 pregnant women with (n=20) and without GDM (n=20) and 20 healthy non-pregnant women. Data as maternal age, gestational age, and body mass index (BMI) of participants were recorded and serum samples were collected. Serum GPER1 levels were measured by enzyme-linked immunosorbent assays. RESULTS Serum GPER1 level was significantly higher in GDM (p=0.03) and non-pregnant women (p=0.005) than those of normal pregnancy. There was no significant correlation between the serum GPER1 levels age (r=0.18, p=0.34), gestational age (r=-0.22, p=0.47), and BMI (r=0.004, p=0.975). CONCLUSIONS Our results suggest that changes in serum GPER1 levels in pregnancy and GDM may be associated with estrogen. More detailed studies should be conducted to monitor the changes and their interactions in serum sex hormones and serum GPER1 levels during GDM.
Collapse
Affiliation(s)
- Berrak Guven
- Department of Biochemistry, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Yesim Akdemir
- Department of Gynecology and Obstetric, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| |
Collapse
|
33
|
Mulrenin IR, Garcia JE, Fashe MM, Loop MS, Daubert MA, Urrutia RP, Lee CR. The impact of pregnancy on antihypertensive drug metabolism and pharmacokinetics: current status and future directions. Expert Opin Drug Metab Toxicol 2021; 17:1261-1279. [PMID: 34739303 DOI: 10.1080/17425255.2021.2002845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Hypertensive disorders of pregnancy (HDP) are rising in prevalence, and increase risk of adverse maternal and fetal outcomes. Physiologic changes occur during pregnancy that alter drug pharmacokinetics. However, antihypertensive drugs lack pregnancy-specific dosing recommendations due to critical knowledge gaps surrounding the extent of gestational changes in antihypertensive drug pharmacokinetics and underlying mechanisms. AREAS COVERED This review (1) summarizes currently recommended medications and dosing strategies for non-emergent HDP treatment, (2) reviews and synthesizes existing literature identified via a comprehensive Pubmed search evaluating gestational changes in the maternal pharmacokinetics of commonly prescribed HDP drugs (notably labetalol and nifedipine), and (3) offers insight into the metabolism and clearance mechanisms underlying altered HDP drug pharmacokinetics during pregnancy. Remaining knowledge gaps and future research directions are summarized. EXPERT OPINION A series of small pharmacokinetic studies illustrate higher oral clearance of labetalol and nifedipine during pregnancy. Pharmacokinetic modeling and preclinical studies suggest these effects are likely due to pregnancy-associated increases in hepatic UGT1A1- and CYP3A4-mediated first-pass metabolism and lower bioavailability. Accordingly, higher and/or more frequent doses may be needed to lower blood pressure during pregnancy. Future research is needed to address various evidence gaps and inform the development of more precise antihypertensive drug dosing strategies.
Collapse
Affiliation(s)
- Ian R Mulrenin
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Julian E Garcia
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Muluneh M Fashe
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew Shane Loop
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Melissa A Daubert
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC
| | - Rachel Peragallo Urrutia
- Division of General Obstetrics and Gynecology, Department of Obstetrics and Gynecology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
34
|
Liu C, Wei P, Li J. The thickness changes of retina in high myopia patients during the third trimester of pregnancy: a pilot study. BMC Ophthalmol 2021; 21:382. [PMID: 34706696 PMCID: PMC8549153 DOI: 10.1186/s12886-021-02137-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/28/2021] [Indexed: 12/04/2022] Open
Abstract
Objectives To observe and compare the difference in retinal thickness using optical coherence tomography (OCT) between patients with high myopia (HM) during the third trimester of pregnancy and age-matched HM non-pregnant women. Methods A case-control study. A total of 39 eyes from 39 HM women in the third trimester (study group) and 50 eyes of 50 age-matched non-pregnant women with HM (control group) were included. All subjects underwent SD-OCT examination. The built-in software was used to measure the retinal thickness in macular region. The data from two groups were compared using independent-samples t test. Results Among the 89 subjects in this study, the mean gestational age of the study group was 35.09 ± 2.44 weeks, and the average age was 32.24 ± 3.75 years. The average age of the control group was 34.04 ± 7.19 years old. Compared with the control group, the average thickness of parafoveal area, and the average thickness of parafoveal superior, inferior, temporal quadrants of the superficial retina and the average thickness of the foveal and parafoveal of the superficial retina were significantly decreased in the study group (P < 0.05). Compared with the control group, the average thickness of all quadrants of the retina in the parafoveal area except the nasal quadrant were significantly decreased in the study group (P < 0.05). Conclusions In this observational study, the retinal thickness of patients with high myopia during the third trimester of pregnancy was thinner than that of non-pregnant women with age-matched high myopia.
Collapse
Affiliation(s)
- Chenchen Liu
- Department of Ophthalmology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Puying Wei
- Department of Ophthalmology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Jun Li
- Department of Ophthalmology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
35
|
Sundararajan A, Vora K, Saiyed S, Natesan S. Comparative Profiling of Salivary Cortisol and Salivary DHEA-S Among Healthy Pregnant and Non-Pregnant Women. Horm Metab Res 2021; 53:602-607. [PMID: 34496410 DOI: 10.1055/a-1551-3722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
During pregnancy, circulatory cortisol levels increase, remaining steady over the second-third trimester. In contrast, profile of salivary cortisol during pregnancy is debatable, more influenced by factors like time of sample collection in the day. Circulatory DHEA-S decrease by at least 50% over the second-third trimester of pregnancy. However, profile of salivary DHEA-S is unclear. Objective was to determine changes in salivary cortisol and DHEA-S in healthy pregnant women, compared to non-pregnant women during late morning-early afternoon sampling to avoid fluctuations associated with other times. Pregnant women in their second-third trimester prospectively (n=500) and non-pregnant women (n=133) were enrolled in study with informed consent. Live birth outcome with no pregnancy complications and≥2.5 Kg infant birth weight were included. Concentrations of salivary cortisol and DHEA-S were determined through ELISA assays. Compared to non-pregnant women, pregnant women demonstrated significant increases in salivary cortisol [median (interquartile range)=4.2 (5.1) nmol/l vs. 17.2 (13.9) nmol/l, p<0.001] and salivary DHEA-S median (interquartile range)=2.7 (2.9) nmol/l vs. 3.8 (3.2) nmol/l, p<0.001). Consistently, quartile scores representing higher levels of salivary cortisol and DHEA-S concentrations demonstrated significant association with pregnancy. Quartile scores representing higher salivary cortisol/DHEA-S ratio demonstrated significant association with pregnancy. Study suggests the indicated time range of saliva sampling might best parallel the established profile of circulatory cortisol in pregnant women. However, unlike cortisol, study indicates that the salivary DHEA-S profile is distinct from the well-known profile of circulatory DHEA-S during pregnancy. A combinatorial approach involving both salivary and circulatory compartments could provide comprehensive picture of DHEA-S and hypothalamus-pituitary-adrenal axis during pregnancy.
Collapse
Affiliation(s)
| | - Kranti Vora
- Indian Institute of Public Health Gandhinagar, Lekawada, Gandhinagar, India
- University of Canberra, Bruce, Australia
| | - Shahin Saiyed
- Indian Institute of Public Health Gandhinagar, Lekawada, Gandhinagar, India
| | | |
Collapse
|
36
|
Rodrigues AD, van Dyk M, Sorich MJ, Fahmy A, Useckaite Z, Newman LA, Kapetas AJ, Mounzer R, Wood LS, Johnson JG, Rowland A. Exploring the Use of Serum-Derived Small Extracellular Vesicles as Liquid Biopsy to Study the Induction of Hepatic Cytochromes P450 and Organic Anion Transporting Polypeptides. Clin Pharmacol Ther 2021; 110:248-258. [PMID: 33792897 DOI: 10.1002/cpt.2244] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/17/2021] [Indexed: 12/14/2022]
Abstract
Liver-derived small extracellular vesicles (sEVs), prepared from small sets of banked serum samples using a novel two-step protocol, were deployed as liquid biopsy to study the induction of cytochromes P450 (CYP3A4, CYP3A5, and CYP2D6) and organic anion transporting polypeptides (OATP1B1 and OATP1B3) during pregnancy (nonpregnant (T0), first, second, and third (T3) trimester women; N = 3 each) and after administration of rifampicin (RIF) to healthy male subjects. Proteomic analysis revealed induction (mean fold-increase, 90% confidence interval) of sEV CYP3A4 after RIF 300 mg × 7 days (3.5, 95% CI = 2.5-4.5, N = 4, P = 0.029) and 600 mg × 14 days (3.7, 95% CI = 2.1-6.0, N = 5, P = 0.018) consistent with the mean oral midazolam area under the plasma concentration time curve (AUC) ratio in the same subjects (0.28, 95% CI = 0.22-0.34, P < 0.0001; and 0.17, 95% CI = 0.13-0.22, P < 0.0001). Compared with CYP3A4, liver sEV CYP3A5 protein (subjects genotyped CYP3A5*1/*3) was weakly induced (≤ 1.5-fold). It was also possible to measure liver sEV-catalyzed dextromethorphan (DEX) O-demethylation to dextrorphan (DXO), correlated with sEV CYP2D6 expression (r = 0.917, P = 0.0001; N = 10) and 3-hour plasma DXO-to-DEX concentration ratio (r = 0.843, P = 0.002, N = 10), and show that CYP2D6 was not induced by RIF. Nonparametric analysis of liver sEV revealed significantly higher CYP3A4 (3.2-fold, P = 0.003) and CYP2D6 (3.7-fold, P = 0.03) protein expression in T3 vs. T0 women. In contrast, expression of both OATPs in liver sEV was unaltered by RIF administration and pregnancy.
Collapse
Affiliation(s)
- A David Rodrigues
- ADME Sciences, Medicine Design, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut, USA
| | - Madelé van Dyk
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Michael J Sorich
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Alia Fahmy
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Zivile Useckaite
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lauren A Newman
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Asha J Kapetas
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Reham Mounzer
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Linda S Wood
- Pharmacogenomics, Precision Medicine, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut, USA
| | - Jillian G Johnson
- Pharmacogenomics, Precision Medicine, Worldwide Research & Development, Pfizer Inc., Groton, Connecticut, USA
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
37
|
Morya AK, Gogia S, Gupta A, Prakash S, Solanki K, Naidu AD. Motherhood: What every ophthalmologist needs to know. Indian J Ophthalmol 2021; 68:1526-1532. [PMID: 32709768 PMCID: PMC7640830 DOI: 10.4103/ijo.ijo_2033_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The aim of this review article is to summarize the available literature on physiologic and pathologic ocular changes during pregnancy and the effect of diseases in pregnancy. A literature search was conducted using PUBMED, MEDLINE, and Cochrane library in English. In addition, the cited references in the published articles were manually reviewed for the relevant results. Pregnancy encompasses a multitude of changes in all body systems, including the visual system of the female. The changes can be physiological, i.e., changes occurring in the lids and adnexa, cornea, conjunctiva, changes in tear film composition and intraocular pressure, retina, choroid, and visual field. Pathological changes in a pregnant woman's eye include changes related to preeclampsia and eclampsia, central serous chorioretinopathy, retinal artery or vein occlusions, and disseminated intravascular coagulation. Preexisting diseases like diabetic retinopathy, Graves' disease, idiopathic intracranial hypertension, various inflammatory conditions can undergo changes in their course during pregnancy. Ophthalmic medications can have an effect on both mother and the baby and hence should be used cautiously. In addition, intrauterine infections play a major role in causing inflammation in the eye of the baby. Hence, vaccination of the mother prior to pregnancy plays an important role in preventing intrauterine infections in the neonate. A regular eye examination in the perinatal period plays a vital role in recognizing ophthalmic pathologies which might require a prompt medical intervention. Pathological ocular diseases should be discriminated from physiologic changes to establish an individualized treatment or preventive plan. This approach to ocular benefits of treatment to the mother should always weigh against the potential harm to the fetus.
Collapse
Affiliation(s)
- Arvind K Morya
- Department of Ophthalmology, AIIMS, Jodhpur, Rajasthan, India
| | - Sonalika Gogia
- Department of Ophthalmology, AIIMS, Jodhpur, Rajasthan, India
| | - Arushi Gupta
- Department of Ophthalmology, AIIMS, Jodhpur, Rajasthan, India
| | - Sujeet Prakash
- Department of Ophthalmology, AIIMS, Jodhpur, Rajasthan, India
| | - Kanchan Solanki
- Department of Ophthalmology, AIIMS, Jodhpur, Rajasthan, India
| | | |
Collapse
|
38
|
Sex Differences in the Exocrine Pancreas and Associated Diseases. Cell Mol Gastroenterol Hepatol 2021; 12:427-441. [PMID: 33895424 PMCID: PMC8255941 DOI: 10.1016/j.jcmgh.2021.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 04/08/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022]
Abstract
Differences in pancreatic anatomy, size, and function exist in men and women. The anatomical differences could contribute to the increase in complications associated with pancreatic surgery in women. Although diagnostic criteria for pancreatitis are the same in men and women, major sex differences in etiology are reported. Alcohol and tobacco predominate in men, whereas idiopathic and obstructive etiologies predominate in women. Circulating levels of estrogens, progesterone, and androgens contribute significantly to overall health outcomes; premenopausal women have lower prevalence of cardiovascular and pancreatic diseases suggesting protective effects of estrogens, whereas androgens promote growth of normal and cancerous cells. Sex chromosomes and gonadal and nongonadal hormones together determine an individual's sex, which is distinct from gender or gender identity. Human pancreatic disease etiology, outcomes, and sex-specific mechanisms are largely unknown. In rodents of both sexes, glucocorticoids and estrogens from the adrenal glands influence pancreatic secretion and acinar cell zymogen granule numbers. Lack of corticotropin-releasing factor receptor 2 function, a G protein-coupled receptor whose expression is regulated by both estrogens and glucocorticoids, causes sex-specific changes in pancreatic histopathology, zymogen granule numbers, and endoplasmic reticulum ultrastructure changes in acute pancreatitis model. Here, we review existing literature on sex differences in the normal exocrine pancreas and mechanisms that operate at homeostasis and diseased states in both sexes. Finally, we review pregnancy-related pancreatic diseases and discuss the effects of sex differences on proposed treatments in pancreatic disease.
Collapse
|
39
|
Khatri R, Fallon JK, Sykes C, Kulick N, Rementer RJB, Miner TA, Schauer AP, Kashuba ADM, Boggess KA, Brouwer KLR, Smith PC, Lee CR. Pregnancy-Related Hormones Increase UGT1A1-Mediated Labetalol Metabolism in Human Hepatocytes. Front Pharmacol 2021; 12:655320. [PMID: 33995076 PMCID: PMC8115026 DOI: 10.3389/fphar.2021.655320] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/11/2021] [Indexed: 11/25/2022] Open
Abstract
Pregnancy-related hormones (PRH) are recognized as important regulators of hepatic cytochrome P450 enzyme expression and function. However, the impact of PRH on the hepatic expression and function of uridine diphosphate glucuronosyltransferases (UGTs) remains unclear. Using primary human hepatocytes, we evaluated the effect of PRH exposure on mRNA levels and protein concentrations of UGT1A1, UGT2B7, and other key UGT enzymes, and on the metabolism of labetalol (a UGT1A1 and UGT2B7 substrate commonly prescribed to treat hypertensive disorders of pregnancy). Sandwich-cultured human hepatocytes (SCHH) from female donors were exposed to the PRH estradiol, estriol, estetrol, progesterone, and cortisol individually or in combination. We quantified protein concentrations of UGT1A1, UGT2B7, and four additional UGT1A isoforms in SCHH membrane fractions and evaluated the metabolism of labetalol to its glucuronide metabolites in SCHH. PRH exposure increased mRNA levels and protein concentrations of UGT1A1 and UGT1A4 in SCHH. PRH exposure also significantly increased labetalol metabolism to its UGT1A1-derived glucuronide metabolite in a concentration-dependent manner, which positively correlated with PRH-induced changes in UGT1A1 protein concentrations. In contrast, PRH did not alter UGT2B7 mRNA levels or protein concentrations in SCHH, and formation of the UGT2B7-derived labetalol glucuronide metabolite was decreased following PRH exposure. Our findings demonstrate that PRH alter expression and function of UGT proteins in an isoform-specific manner and increase UGT1A1-mediated labetalol metabolism in human hepatocytes by inducing UGT1A1 protein concentrations. These results provide mechanistic insight into the increases in labetalol clearance observed in pregnant individuals.
Collapse
Affiliation(s)
- Raju Khatri
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - John K. Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Craig Sykes
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Natasha Kulick
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Rebecca J. B. Rementer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Taryn A. Miner
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Amanda P. Schauer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Angela D. M. Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kim A. Boggess
- Department of Obstetrics and Gynecology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kim L. R. Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Philip C. Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
40
|
Banker M, Puttabyatappa M, O’Day P, Goodrich JM, Kelley AS, Domino SE, Smith YR, Dolinoy DC, Song PXK, Auchus RJ, Padmanabhan V. Association of Maternal-Neonatal Steroids With Early Pregnancy Endocrine Disrupting Chemicals and Pregnancy Outcomes. J Clin Endocrinol Metab 2021; 106:665-687. [PMID: 33280001 PMCID: PMC7947779 DOI: 10.1210/clinem/dgaa909] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/12/2022]
Abstract
CONTEXT Steroids play an important role in fetal development and parturition. Gestational exposures to endocrine-disrupting chemicals (EDCs) affect steroidal milieu and pregnancy outcomes, raising the possibility of steroids serving as biomarkers. Most studies have not addressed the impact of EDC mixtures, which are reflective of real life scenarios. OBJECTIVE Assess the association of maternal and neonatal steroids with pregnancy outcomes and early pregnancy EDC levels. DESIGN Prospective analysis of mother-infant dyads. SETTING University hospital. PARTICIPANTS 121 mother-infant dyads. MAIN OUTCOME MEASURES The associations of maternal and neonatal steroidal hormones from 121 dyads with pregnancy outcomes, the associations of first trimester EDCs individually and as mixtures with maternal and neonatal steroids in a subset of 56 dyads and the influence of body mass index (BMI), age, and offspring sex in modulating the EDC associations with steroids were determined. RESULTS Steroid-specific positive or negative associations with pregnancy measures were evident; many maternal first trimester EDCs were negatively associated with estrogens and positively with androgen/estrogen ratios; EDC-steroid associations were influenced by maternal age, pre-pregnancy BMI, and fetal sex; and EDCs individually and as mixtures showed direct and inverse fetal sex-dependent associations with maternal and neonatal steroids. CONCLUSIONS This proof-of-concept study indicates association of steroids with pregnancy outcomes depending on maternal age, prepregnancy BMI, and fetal sex, with the effects of EDCs differing when considered individually or as mixtures. These findings suggest that steroidal hormonal measures have potential to serve as biomarkers of impact of EDC exposures and pregnancy outcome.
Collapse
Affiliation(s)
- Margaret Banker
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | | | - Patrick O’Day
- Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology, & Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Jaclyn M Goodrich
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Angela S Kelley
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Steven E Domino
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Yolanda R Smith
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Peter X K Song
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Richard J Auchus
- Departments of Pharmacology and Internal Medicine, Division of Metabolism, Endocrinology, & Diabetes, University of Michigan, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
41
|
Vuppaladhadiam L, Lager J, Fiehn O, Weiss S, Chesney M, Hasdemir B, Bhargava A. Human Placenta Buffers the Fetus from Adverse Effects of Perceived Maternal Stress. Cells 2021; 10:cells10020379. [PMID: 33673157 PMCID: PMC7918582 DOI: 10.3390/cells10020379] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 11/16/2022] Open
Abstract
Maternal stress during pregnancy is linked to several negative birth outcomes. The placenta, a unique pregnancy-specific organ, not only nourishes and protects the fetus but is also the major source of progesterone and estrogens. As the placenta becomes the primary source of maternal progesterone (P4) and estradiol between 6-9 weeks of gestation, and these hormones are critical for maintaining pregnancy, maternal stress may modulate levels of these steroids to impact birth outcomes. The objective was to test whether maternal perceived stress crosses the placental barrier to modulate fetal steroids, including cortisol, which is a downstream indicator of maternal hypothalamic-pituitary-adrenal (HPA) axis regulation and is associated with negative fetal outcomes. Nulliparous women, 18 years or older, with no known history of adrenal or endocrine illness were recruited during their third trimester of pregnancy at the University of California San Francisco (UCSF) Mission Bay hospital obstetrics clinics. Simultaneous measurement of 10 steroid metabolites in maternal (plasma and hair) and fetal (cord blood and placenta) samples was performed using tandem mass spectrometry along with assessment of the perceived stress score and sociodemographic status. While the maternal perceived stress score (PSS) and sociodemographic status were positively associated with each other and each with the body mass index (BMI) (r = 0.73, p = 0.0008; r = 0.48, p = 0.05; r = 0.59, p = 0.014, respectively), PSS did not correlate with maternal or fetal cortisol, cortisone levels, or fetal birth weight. Regardless of maternal PSS or BMI, fetal steroid levels remained stable and unaffected. Progesterone was the only steroid analyte quantifiable in maternal hair and correlated positively with PSS (r = 0.964, p = 0.003), whereas cord estradiol was negatively associated with PSS (r = -0.94, p = 0.017). In conclusion, hair progesterone might serve as a better marker of maternal stress than cortisol or cortisone and maternal PSS negatively impacts fetal estradiol levels. Findings have implications for improved biomarkers of stress and targets for future research to identify factors that buffer the fetus from adverse effects of maternal stress.
Collapse
Affiliation(s)
- Lahari Vuppaladhadiam
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA; (J.L.); (B.H.)
| | - Jeannette Lager
- Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA; (J.L.); (B.H.)
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA 95616, USA;
| | - Sandra Weiss
- Department of Community Health Systems, Stress and Depression Research Lab, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Margaret Chesney
- The Osher Center for Integrative Medicine, University of California San Francisco, San Francisco, CA 94143, USA;
| | - Burcu Hasdemir
- Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA; (J.L.); (B.H.)
- NIH West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA 95616, USA;
| | - Aditi Bhargava
- Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA 94143, USA;
- Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA; (J.L.); (B.H.)
- NIH West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA 95616, USA;
- Correspondence: ; Tel.: +1-415-502-8453
| |
Collapse
|
42
|
Liang L, Rasmussen MLH, Piening B, Shen X, Chen S, Röst H, Snyder JK, Tibshirani R, Skotte L, Lee NC, Contrepois K, Feenstra B, Zackriah H, Snyder M, Melbye M. Metabolic Dynamics and Prediction of Gestational Age and Time to Delivery in Pregnant Women. Cell 2021; 181:1680-1692.e15. [PMID: 32589958 PMCID: PMC7327522 DOI: 10.1016/j.cell.2020.05.002] [Citation(s) in RCA: 145] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 03/11/2020] [Accepted: 04/29/2020] [Indexed: 01/10/2023]
Abstract
Metabolism during pregnancy is a dynamic and precisely programmed process, the failure of which can bring devastating consequences to the mother and fetus. To define a high-resolution temporal profile of metabolites during healthy pregnancy, we analyzed the untargeted metabolome of 784 weekly blood samples from 30 pregnant women. Broad changes and a highly choreographed profile were revealed: 4,995 metabolic features (of 9,651 total), 460 annotated compounds (of 687 total), and 34 human metabolic pathways (of 48 total) were significantly changed during pregnancy. Using linear models, we built a metabolic clock with five metabolites that time gestational age in high accordance with ultrasound (R = 0.92). Furthermore, two to three metabolites can identify when labor occurs (time to delivery within two, four, and eight weeks, AUROC ≥ 0.85). Our study represents a weekly characterization of the human pregnancy metabolome, providing a high-resolution landscape for understanding pregnancy with potential clinical utilities. Weekly metabolome of maternal blood changes dynamically through healthy pregnancy A metabolic clock of five blood metabolites accurately predicts gestational age Two to three metabolites identify labor onset within two, four, and eight weeks Women with metabolic clocks that outpaced ultrasound evaluation tend to deliver earlier
Collapse
Affiliation(s)
- Liang Liang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Brian Piening
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xiaotao Shen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannes Röst
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John K Snyder
- Department of Chemistry and the Chemical Instrumentation Center, Boston University, Boston, Massachusetts 02215, USA
| | - Robert Tibshirani
- Department of Statistics and Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Line Skotte
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, 2300, Denmark
| | - Norman Cy Lee
- Department of Chemistry and the Chemical Instrumentation Center, Boston University, Boston, Massachusetts 02215, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bjarke Feenstra
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, 2300, Denmark
| | - Hanyah Zackriah
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Mads Melbye
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, 2300, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, 2200, Denmark; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
43
|
Khatri R, Kulick N, Rementer RJB, Fallon JK, Sykes C, Schauer AP, Malinen MM, Mosedale M, Watkins PB, Kashuba ADM, Boggess KA, Smith PC, Brouwer KLR, Lee CR. Pregnancy-Related Hormones Increase Nifedipine Metabolism in Human Hepatocytes by Inducing CYP3A4 Expression. J Pharm Sci 2021; 110:412-421. [PMID: 32931777 PMCID: PMC7750305 DOI: 10.1016/j.xphs.2020.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022]
Abstract
Pregnancy-related hormones (PRH) have emerged as key regulators of hepatic cytochrome P450 (CYP) enzyme expression and function. The impact of PRH on protein levels of CYP3A4 and other key CYP enzymes, and the metabolism of nifedipine (a CYP3A4 substrate commonly prescribed during pregnancy), was evaluated in primary human hepatocytes. Sandwich-cultured human hepatocytes (SCHH) from female donors were exposed to PRH (estradiol, estriol, estetrol, progesterone, and cortisol), individually or in combination as a cocktail. Absolute protein concentrations of twelve CYP isoforms in SCHH membrane fractions were quantified by nanoLC-MS/MS, and metabolism of nifedipine to dehydronifedipine in SCHH was evaluated. PRH significantly increased CYP3A4 protein concentrations and nifedipine metabolism to dehydronifedipine in a concentration-dependent manner. CYP3A4 mRNA levels in hepatocyte-derived exosomes positively correlated with CYP3A4 protein levels and dehydronifedipine formation in SCHH. PRH also increased CYP2B6, CYP2C8 and CYP2A6 levels. Our findings demonstrate that PRH increase nifedipine metabolism in SCHH by inducing CYP3A4 expression and alter expression of other key CYP proteins in an isoform-specific manner, and suggest that hepatocyte-derived exosomes warrant further investigation as biomarkers of hepatic CYP3A4 metabolism. Together, these results offer mechanistic insight into the increases in nifedipine metabolism and clearance observed in pregnant women.
Collapse
Affiliation(s)
- Raju Khatri
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Natasha Kulick
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca J B Rementer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John K Fallon
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig Sykes
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amanda P Schauer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melina M Malinen
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Merrie Mosedale
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Paul B Watkins
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angela D M Kashuba
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kim A Boggess
- Department of Obstetrics and Gynecology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Philip C Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kim L R Brouwer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Craig R Lee
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
44
|
McCoy SS, Giri J, Das R, Paul PK, Pennati A, Parker M, Liang Y, Galipeau J. Minor salivary gland mesenchymal stromal cells derived from patients with Sjӧgren's syndrome deploy intact immune plasticity. Cytotherapy 2020; 23:301-310. [PMID: 33262072 DOI: 10.1016/j.jcyt.2020.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 09/16/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSCs) provide minor salivary glands (MSGs) with support and niche cells for epithelial glandular tissue. Little is known about resident MSG-derived MSCs (MSG-MSCs) in primary Sjӧgren's syndrome (PSS). The authors' objective is to define the immunobiology of endogenous PSS MSG-MSCs. METHODS Using culture-adapted MSG-MSCs isolated from consenting PSS subjects (n = 13), the authors performed in vitro interrogation of PSS MSG-MSC immunobiology and global gene expression compared with controls. To this end, the authors performed phenotypic and immune functional analysis of indoleamine 2,3-dioxygenase (IDO), programmed death ligand 1 (PD-L1) and intercellular adhesion marker 1 (ICAM-1) before and after interferon γ (IFNγ) licensing as well as the effect of MSG-MSCs on T-cell proliferation. Considering the female predominance of PSS, the authors also addressed the influence of 17-β-estradiol on estrogen receptor α-positive-related MSC function. RESULTS The authors found that MSG-MSCs deployed normal immune regulatory functionality after IFNγ stimulation, as demonstrated by increased protein-level expression of IDO, PD-L1 and ICAM-1. The authors also found that MSG-MSCs suppressed T-cell proliferation in a dose-dependent manner independent of 17-β-estradiol exposure. Gene ontology and pathway analysis highlighted extracellular matrix deposition as a possible difference between PSS and control MSG-MSCs. MSG-MSCs demonstrated increased α-smooth muscle actin expression in PSS, indicating a partial myofibroblast-like adaptation. CONCLUSIONS These findings establish similar immune regulatory function of MSG-MSCs in both PSS and control patients, precluding intrinsic MSC immune regulatory defects in PSS. PSS MSG-MSCs show a partial imprinted myofibroblast-like phenotype that may arise in the setting of chronic inflammation, providing a plausible etiology for PSS-related glandular fibrosis.
Collapse
Affiliation(s)
- Sara S McCoy
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| | - Jayeeta Giri
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Rahul Das
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Pradyut K Paul
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Andrea Pennati
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| | - Maxwell Parker
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Yun Liang
- Department of Medical Biology and Immunology, University of Wisconsin, Madison, Wisconsin, USA
| | - Jacques Galipeau
- Department of Medicine, University of Wisconsin Carbone Comprehensive Cancer Center, University of Wisconsin, Madison, Wisconsin, USA
| |
Collapse
|
45
|
Swanson LM, Kalmbach DA, Raglan GB, O’Brien LM. Perinatal Insomnia and Mental Health: a Review of Recent Literature. Curr Psychiatry Rep 2020; 22:73. [PMID: 33104878 PMCID: PMC9109228 DOI: 10.1007/s11920-020-01198-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The perinatal period is a time of high risk for insomnia and mental health conditions. The purpose of this review is to critically examine the most recent literature on perinatal insomnia, focusing on unique features of this period which may confer specific risk, associations with depression and anxiety, and emerging work on perinatal insomnia treatment. RECENT FINDINGS A majority of perinatal women experience insomnia, which may persist for years, and is associated with depression and anxiety. Novel risk factors include personality characteristics, nocturnal perinatal-focused rumination, and obesity. Mindfulness and physical activity may be protective. Cognitive-behavioral therapy for insomnia is an effective treatment. Perinatal insomnia is exceedingly common, perhaps due to factors unique to this period. Although closely linked to perinatal mental health, more work is needed to establish causality. Future work is also needed to establish the role of racial disparities, tailor treatments, and determine whether insomnia treatment improves perinatal mental health.
Collapse
Affiliation(s)
- Leslie M. Swanson
- Department of Psychiatry, University of Michigan, Ann Arbor, MI;,Corresponding Author: Leslie M. Swanson, Department of Psychiatry, University of Michigan, 4250 Plymouth Rd, Ann Arbor, MI 48109,
| | - David A. Kalmbach
- Sleep Disorders and Research Center, Henry Ford Hospital, Detroit, MI
| | - Greta B. Raglan
- Department of Psychiatry, University of Michigan, Ann Arbor, MI
| | - Louise M. O’Brien
- Department of Neurology, Sleep Disorders Center, University of Michigan, Ann Arbor, MI
| |
Collapse
|
46
|
Qiu W, Hodges TE, Clark EL, Blankers SA, Galea LAM. Perinatal depression: Heterogeneity of disease and in animal models. Front Neuroendocrinol 2020; 59:100854. [PMID: 32750403 DOI: 10.1016/j.yfrne.2020.100854] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/14/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022]
Abstract
Perinatal depression (PND) can have either an antepartum or postpartum onset. Although the greatest risk factor for PND is previous depression history,de novoPND occurs with the majority of cases occurring in the postpartum. Timing of depression can impact etiology, prognosis, and response to treatment. Thus, it is crucial to study the impact of the heterogeneity of PND for better health outcomes. In this review, we outline the differences between antepartum and postpartum depression onset of PND. We discuss maternal physiological changes that differ between pregnancy and postpartum and how these may differentially impact depression susceptibility. We highlight changes in the maternal steroid and peptide hormone levels, immune signalling, serotonergic tone, metabolic factors, brain morphology, and the gut microbiome. Finally, we argue that studying the heterogeneity of PND in clinical and preclinical models can lead to improved knowledge of disease etiopathology and treatment outcomes.
Collapse
Affiliation(s)
- Wansu Qiu
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Travis E Hodges
- Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Emily L Clark
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Samantha A Blankers
- Graduate Program in Neuroscience, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada
| | - Liisa A M Galea
- Graduate Program in Neuroscience, University of British Columbia, Canada; Department of Psychology, University of British Columbia, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Canada.
| |
Collapse
|
47
|
Abstract
OBJECTIVE Prevalence of Alzheimer's disease (AD) is higher for women, possibly influenced by sex-dependent effects of the estrogen. We examined the association between estrogen and cognitive decline in over 2,000 older adult women in a 12-year population-based study in Cache County, Utah. METHODS The baseline sample included 2,114 women (mean age = 74.94 y, SD = 6.71) who were dementia-free at baseline and completed a women's health questionnaire, asking questions regarding reproductive history and hormone therapy (HT). Endogenous estrogen exposure (EEE) was calculated taking the reproductive window (age at menarche to age at menopause), adjusted for pregnancy and breastfeeding. HT variables included duration of use, HT type (unopposed; opposed), and time of HT initiation. A modified version of the Mini-Mental State Examination (3MS) was administered at four triennial waves to assess cognitive status. Linear mixed-effects models examined the relationship between estrogen exposure and 3MS score over time. RESULTS EEE was positively associated with cognitive status (β = 0.03, P = 0.054). In addition, longer duration of HT use was positively associated with cognitive status (β = 0.02, P = 0.046) and interacted with age; older women had greater benefit compared with younger women. The timing of HT initiation was significantly associated with 3MS (β = 0.55, P = 0.048), with higher scores for women who initiated HT within 5 years of menopause compared with those initiating HT 6-or-more years later. CONCLUSIONS Our results suggest that longer EEE and HT use, especially in older women, are associated with higher cognitive status in late life.
Collapse
|
48
|
Abstract
Hepatic drug metabolism is a major route of drug elimination, mediated by multiple drug-metabolizing enzymes. Any changes in the rate and extent of hepatic drug metabolism can lead to altered drug efficacy or toxicity. Accumulating clinical evidence indicates that pregnancy is accompanied by changes in hepatic drug metabolism. In this article, we discuss in vitro and in vivo tools used to study the mechanisms underlying the altered drug metabolism during pregnancy, focusing on primary hepatocyte culture, transgenic animal models, and use of probe drugs to assess change in enzymatic activity. The information obtained from these studies has enabled prediction of clinical PK changes for a given drug in pregnant women.
Collapse
Affiliation(s)
- Hyunyoung Jeong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, 900 S. Ashland Ave, Chicago, IL 60607, United States.
| | - Catherine S. Stika
- Department of Obstetrics and Gynecology, Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
49
|
Kızıltunç PB, Varlı B, Büyüktepe TÇ, Atilla H. Ocular vascular changes during pregnancy: an optical coherence tomography angiography study. Graefes Arch Clin Exp Ophthalmol 2019; 258:395-401. [PMID: 31754828 DOI: 10.1007/s00417-019-04541-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/29/2019] [Accepted: 11/07/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Pregnancy is a period presenting with many physiological adaptation mechanisms. One of the structures in which these mechanisms are observed is ocular tissues. The cornea, lacrimal and meibomian glands, and chorioretinal complex are all among the structures affected by changes during pregnancy. In this study we aimed to evaluate the macular and optic disc vessel density (VD) changes by Optical Coherence Tomography Angiography (OCTA) imaging in pregnancy. METHODS A total of 248 eyes from 124 pregnant women and 80 eyes from 40 healthy control women were involved. Vessel densities of macula were evaluated for superficial capillary plexus (SCP) and deep capillary plexus (DCP) in whole macula, foveal, parafoveal and perifoveal region. Peripapillary and whole optic disc VDs were also evaluated. Vessel densities of macula and optic disc were compared between control individuals and pregnant women. Vessel densities in different trimesters were also evaluated. RESULTS Modest but significant differences in VDs of whole macula of SCP and DCP were observed in pregnancy group. Additionally, perifoveal and parafoveal region of SCP, whole disc and radial peripapillary capillary VD were significantly higher in pregnancy group. There was no correlation between VD ratios of macula and optic disc and pregnancy weeks and trimesters. CONCLUSIONS This is the first study focusing on the OCTA parameters in pregnant individuals. These findings suggest that physiological changes during pregnancy are not limited to the cornea, eyelids and the choroid but also to the retinal and optic disc vasculature.
Collapse
Affiliation(s)
- Pınar Bingöl Kızıltunç
- Department of Ophthalmology, Ankara University School of Medicine, Vehbi Koç Eye Bank, Mamak, Ankara, Turkey.
| | - Bulut Varlı
- Department of Obstetrics and Gynecology, Ankara University School of Medicine, Ankara, Turkey
| | - Tuna Çelik Büyüktepe
- Department of Ophthalmology, Ankara University School of Medicine, Vehbi Koç Eye Bank, Mamak, Ankara, Turkey
| | - Huban Atilla
- Department of Ophthalmology, Ankara University School of Medicine, Vehbi Koç Eye Bank, Mamak, Ankara, Turkey
| |
Collapse
|
50
|
Ravi M, Stevens JS, Michopoulos V. Neuroendocrine pathways underlying risk and resilience to PTSD in women. Front Neuroendocrinol 2019; 55:100790. [PMID: 31542288 PMCID: PMC6876844 DOI: 10.1016/j.yfrne.2019.100790] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/26/2019] [Accepted: 09/18/2019] [Indexed: 01/23/2023]
Abstract
Women are twice as likely than men to suffer from posttraumatic stress disorder (PTSD). While women have increased exposure to traumatic events of many types and have greater prevalence of comorbid psychiatric disorders compared to men, these differences do not account for the overall sex difference in the prevalence of PTSD. The current review summarizes significant findings that implicate the role of estradiol, progesterone, and allopregnanolone in female risk for PTSD symptoms and dysregulation of fear psychophysiology that is cardinal to PTSD. We also discuss how these steroid hormones influence the stress axis and neural substrates critical for the regulation of fear responses. Understanding the role of ovarian steroid hormones in risk and resilience for trauma-related adverse mental health outcomes across the lifespan in women has important translational, clinical, and intergenerational implications for mitigating the consequences of trauma exposure.
Collapse
Affiliation(s)
- Meghna Ravi
- Emory University Graduate Program in Neuroscience, Atlanta, GA, United States
| | - Jennifer S Stevens
- Emory University School of Medicine, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, United States
| | - Vasiliki Michopoulos
- Emory University School of Medicine, Department of Psychiatry and Behavioral Sciences, Atlanta, GA, United States; Yerkes National Primate Research Center, Atlanta, GA, United States.
| |
Collapse
|