1
|
Singh A, Perez ML, Kirsanov O, Padilla-Banks E, Guardia CM. Autophagy in reproduction and pregnancy-associated diseases. iScience 2024; 27:111268. [PMID: 39628569 PMCID: PMC11613427 DOI: 10.1016/j.isci.2024.111268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
As advantageous as sexual reproduction is during progeny generation, it is also an expensive and treacherous reproductive strategy. The viviparous eukaryote has evolved to survive stress before, during, and after pregnancy. An important and conserved intracellular pathway for the control of metabolic stress is autophagy. The autophagy process occurs in multiple stages through the coordinated action of autophagy-related genes. This review summarizes the evidence that autophagy is an integral component of reproduction. Additionally, we discuss emerging in vitro techniques that will enable cellular and molecular studies of autophagy and its associated pathways in reproduction. Finally, we discuss the role of autophagy in the pathogenesis and progression of several pregnancy-related disorders such as preterm birth, preeclampsia, and intra-uterine growth restriction, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Asmita Singh
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Maira L. Perez
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Oleksandr Kirsanov
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Elizabeth Padilla-Banks
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Carlos M. Guardia
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
2
|
Wu X, Shen J, Liu J, Kang N, Zhang M, Cai X, Zhen X, Yan G, Liu Y, Sun H. Increased EHD1 in trophoblasts causes RSM by activating TGFβ signaling†. Biol Reprod 2024; 111:1235-1248. [PMID: 39012723 DOI: 10.1093/biolre/ioae110] [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: 04/16/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 07/18/2024] Open
Abstract
BACKGROUND Recurrent spontaneous miscarriage is one of the complications during pregnancy. However, the pathogenesis of recurrent spontaneous miscarriage is far from fully elucidated. OBJECTIVE Since the endocytic pathway is crucial for cellular homeostasis, our study aimed to explore the roles of endocytic recycling, especially EH domain containing 1, a member of the endocytic recycling compartment, in recurrent spontaneous miscarriage. STUDY DESIGN We first investigated the expression of the endocytic pathway member EH domain containing 1 in villi from the normal and recurrent spontaneous miscarriage groups. Then, we performed ribonucleic acid sequencing and experiments in villi, HTR8 cells and BeWo cells to determine the mechanisms by which EH domain containing 1-induced recurrent spontaneous miscarriage. Finally, placenta-specific EH domain containing 1-overexpressing mice were generated to investigate the recurrent spontaneous miscarriage phenotype in vivo. RESULTS EH domain containing 1 was expressed in extravillous trophoblasts and syncytiotrophoblast in the villi. Compared with the control group, recurrent spontaneous miscarriage patients expressed higher EH domain containing 1. A high level of EH domain containing 1 decreased proliferation, promoted apoptosis, and reduced the migration and invasion of HTR8 cells by activating the TGFβ receptor 1-SMAD2/3 signaling pathway. The TGFβ receptor 1 antagonist LY3200882 partially reversed the EH domain containing 1 overexpression-induced changes in the cell phenotype. Besides, a high level of EH domain containing 1 also induced abnormal syncytialization, which disturbed maternal-fetal material exchanges. In a mouse model, placenta-specific overexpression of EH domain containing 1 led to the failure of spiral artery remodeling, excessive syncytialization, and miscarriage. CONCLUSIONS Increased expression of EH domain containing 1 impaired the invasion of extravillous trophoblasts mediated by the TGFβ receptor 1-SMAD2/3 signaling pathway and induced abnormal syncytialization of syncytiotrophoblast, which is at least partially responsible for recurrent spontaneous miscarriage.
Collapse
Affiliation(s)
- Xing Wu
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Jiayan Shen
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Jinjin Liu
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Nannan Kang
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Mingshun Zhang
- NHC Key Laboratory of Antibody Technique, Jiangsu Key Laboratory of Pathogen Biology, Department of Immunology, Nanjing Medical University, No. 101 Longmian Avenue, Nanjing 211166, People's Republic of China
| | - Xinyu Cai
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Xin Zhen
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Guijun Yan
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Xuzhou Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Yang Liu
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| | - Haixiang Sun
- Reproductive Medicine Centre, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
- Reproductive Medicine Center, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No. 53 Zhongshan North Road, Nanjing 210008, People's Republic of China
| |
Collapse
|
3
|
da Silva RJ, Cabo LF, Boyle JP. Teratogenic parasites: disease mechanisms and emerging study models. Trends Parasitol 2024; 40:1159-1172. [PMID: 39572326 DOI: 10.1016/j.pt.2024.10.016] [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: 09/23/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 12/07/2024]
Abstract
Congenital infections are a leading preventable cause of pregnancy complications impacting both mother and fetus. Although advancements have been made in understanding various congenital infections, the mechanisms of parasitic infections during pregnancy remain poorly understood. This review covers the global incidence of three parasites capable of congenital transmission - Trypanosoma cruzi, Plasmodium spp., and Toxoplasma gondii - and the state of research into their transplacental transmission strategies. We highlight technological advancements in placental modeling that offer opportunities to reveal how parasites cause gestational pathology. Additionally, we discuss the likelihood that selective adaptation contributed to the evolution of mechanisms that facilitate placental infection. These insights provide a foundation for understanding the progression and pathology of congenital parasitic diseases and identifying future research directions.
Collapse
Affiliation(s)
- Rafaela Jose da Silva
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leah F Cabo
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jon P Boyle
- Department of Biological Sciences, Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Zhou J, Yan P, Ma W, Li J. Cytokine modulation and immunoregulation of uterine NK cells in pregnancy disorders. Cytokine Growth Factor Rev 2024:S1359-6101(24)00095-9. [PMID: 39603954 DOI: 10.1016/j.cytogfr.2024.11.007] [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: 11/11/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Uterine natural killer (uNK) cells play a pivotal role in promoting placental development and supporting maternal-fetal immune tolerance, primarily through cytokine regulation and growth factor production. While the importance of uNK cells in pregnancy is well-established, the mechanisms of their interactions with trophoblasts and contributions to various pregnancy complications remain incompletely understood. This review highlights recent advancements in understanding uNK cell functions, with a focus on cytokine production, growth factor secretion, and receptor-ligand interactions, particularly involving killer immunoglobulin-like receptors (KIR) and human leukocyte antigen-C (HLA-C). We explore how uNK cell dysfunction contributes to pregnancy complications, including preeclampsia, recurrent pregnancy loss, and placenta accreta spectrum (PAS) disorders, emphasizing their roles in immune tolerance and placental health. By detailing the distinct cytokine signaling pathways and functional subtypes of uNK cells, this review provides insights into their regulatory mechanisms essential for pregnancy maintenance. Additionally, we discuss emerging therapeutic strategies targeting uNK-trophoblast interactions and propose future research directions, including the development of non-invasive biomarkers and personalized interventions. This comprehensive review addresses critical knowledge gaps, aiming to advance research in reproductive immunology and guide therapeutic innovations in maternal health.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China.
| | - Ping Yan
- Qingdao City Health Care Center for Cadres, Qingdao, Shandong 266071, China.
| | - Wenxue Ma
- Division of Regenerative Medicine, Department of Medicine, Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA 92093, USA.
| | - Jing Li
- Department of Obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China.
| |
Collapse
|
5
|
Lei D, Chen T, Fan C, Xie Q. Exposure to BaA inhibits trophoblast cell invasion and induces miscarriage by regulating the DEC1/ARHGAP5 axis and promoting ubiquitination-mediated degradation of MMP2. JOURNAL OF HAZARDOUS MATERIALS 2024; 479:135594. [PMID: 39191013 DOI: 10.1016/j.jhazmat.2024.135594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Benz[a]anthracene (BaA), a hazardous polycyclic aromatic hydrocarbon classified by the EPA, is a probable reproductive toxicant. Epidemiological studies suggest that BaA exposure may be a risk factor for recurrent miscarriage (RM). However, the underlying mechanisms are not well understood. This study identified DEC1 as a key gene through RNA-seq and single-cell RNA sequencing analysis. DEC1 expression was found to be downregulated in villous tissues from women with RM and in primary extravillous trophoblasts (EVTs) exposed to BaA. BaA suppressed DEC1 expression by promoting abnormal methylation patterns. Further analysis revealed that ARHGAP5 is a direct target of DEC1 in EVTs, where DEC1 inhibits trophoblast invasion by directly regulating ARHGAP5 transcription. Additionally, BaA destabilized matrix metalloproteinase 2 (MMP2) by activating the aryl hydrocarbon receptor (AhR) and promoting E3 ubiquitin ligase MID1-mediated degradation. In a mouse model, BaA induced miscarriage by modulating the DEC1/ARHGAP5 and MID1/MMP2 axes. Notably, BaA-induced miscarriage in mice was prevented by DEC1 overexpression or MID1 knockdown. These findings indicate that BaA exposure leads to miscarriage by suppressing the DEC1/ARHGAP5 pathway and enhancing the MID1/MMP2 pathway in human EVTs.
Collapse
Affiliation(s)
- Di Lei
- Centre for Reproductive Medicine, Renmin Hospital of Wuhan University, Wuhan 430000, China; Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430000, China
| | - Tingting Chen
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430000, China
| | - Cuifang Fan
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430000, China
| | - Qingzhen Xie
- Centre for Reproductive Medicine, Renmin Hospital of Wuhan University, Wuhan 430000, China.
| |
Collapse
|
6
|
Liu S, Gao M, Zhang X, Wei J, Cui H. FOXP2 overexpression upregulates LAMA4 expression and thereby alleviates preeclampsia by regulating trophoblast behavior. Commun Biol 2024; 7:1427. [PMID: 39487340 PMCID: PMC11530449 DOI: 10.1038/s42003-024-07149-7] [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: 04/11/2024] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
Preeclampsia (PE) is a common pregnancy disorder characterized by hypertension and proteinuria. Trophoblast behavior severely affect PE progression. Transcription factor Forkhead box protein P2 (FOXP2) was involved in cell migration and invasion, but its role in PE progression remains unknown. Laminin subunit alpha 4 (LAMA4) was predicted as a downstream gene of FOXP2 and related to PE. Thus, we supposed that FOXP2 might regulate PE by regulating LAMA4. We found the decreased FOXP2 expression in patients with PE compared with healthy pregnant women. The rat model of PE was induced by L-NAME oral gavage. FOXP2 overexpression lowered systolic and diastolic blood pressure and restored pathological changes of rats with PE. Trophoblasts under the hypoxia/reoxygenation (H/R) treatment were used to mimic PE in vitro. The results revealed that FOXP2 overexpression inhibited apoptosis but promoted migration, invasion, and angiogenesis of H/R-treated trophoblasts. Dual luciferase and chromatin immunoprecipitation-polymerase chain reaction assays confirmed that FOXP2 transcriptionally upregulated the LAMA4 expression in trophoblasts. LAMA4 knockdown reversed the migration and invasion-promoting role of FOXP2 overexpression in trophoblasts with H/R treatment. Collectively, our findings suggest that the FOXP2/LAMA4 axis regulates PE by suppressing trophoblast apoptosis and promoting its migration, invasion, and angiogenesis.
Collapse
Affiliation(s)
- Sishi Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36# Sanhao Street, Shenyang, 110004, China
| | - Man Gao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36# Sanhao Street, Shenyang, 110004, China
| | - Xue Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36# Sanhao Street, Shenyang, 110004, China
| | - Jun Wei
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36# Sanhao Street, Shenyang, 110004, China
| | - Hong Cui
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, 36# Sanhao Street, Shenyang, 110004, China.
| |
Collapse
|
7
|
Gonçalves IM, Afzal M, Kennedy N, Moita A, Lima R, Ostrovidov S, Hori T, Nashimoto Y, Kaji H. Placental microphysiological systems: new advances on promising platforms that mimic the microenvironment of the human placenta. LAB ON A CHIP 2024. [PMID: 39417748 DOI: 10.1039/d4lc00500g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
One of the most complex human physiological processes to study is pregnancy. Standard animal models, as well as two-dimensional models, lack the complexity and biological relevance required to accurately study such a physiological process. Recent studies have focused on the development of three-dimensional models based on microfluidic systems, designated as placental microphysiological systems (PMPSs). PMPS devices provide a model of the placental barrier through culturing relevant cell types in specific arrangements and media to mimic the in vivo environment of the maternal-fetal circulation. Here, recent developments of PMPS models for embryo uterine implantation, preeclampsia evaluation, and toxicological screening are presented. Studies that use bioprinting techniques are also discussed. Lastly, recent developments in endometrium microphysiological systems are reviewed. All these presented models showed their superiority compared to standard models in recapitulating the biological environment seen in vivo. However, several limitations regarding the types of cells and materials used for these systems were also widely reported. Despite the need for further improvements, PMPS models contribute to a better understanding of the biological mechanisms surrounding pregnancy and the respective pathologies.
Collapse
Affiliation(s)
- Inês M Gonçalves
- METRICS, University of Minho, Guimarães, Portugal
- IN+, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Muhammad Afzal
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Nithil Kennedy
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
- Faculty of Medicine, Imperial College London, London, UK
| | - Ana Moita
- IN+, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Centro de Investigação Desenvolvimento e Inovação da Academia Militar, Academia Militar, Instituto Universitário Militar, Rua Gomes Freire, 1169-203, Lisboa, Portugal
| | - Rui Lima
- METRICS, University of Minho, Guimarães, Portugal
- CEFT, Faculty of Engineering of the University of Porto, Porto, Portugal
- ALiCE, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Serge Ostrovidov
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Takeshi Hori
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Yuji Nashimoto
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
| | - Hirokazu Kaji
- Department of Diagnostic and Therapeutic Systems Engineering, Laboratory for Biomaterials and Bioengineering, Institute of Integrated Research (IIR), Institute of Science Tokyo, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan.
- Research Center for Autonomous Systems Materialogy (ASMat), Institute of Integrated Research (IIR), Institute of Science Tokyo, Japan
| |
Collapse
|
8
|
Huang H, Gao S, Bao M. Exploring Mechanical Forces Shaping Self-Organization and Morphogenesis During Early Embryo Development. Annu Rev Cell Dev Biol 2024; 40:75-96. [PMID: 38608312 DOI: 10.1146/annurev-cellbio-120123-105748] [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: 04/14/2024]
Abstract
Embryonic development is a dynamic process orchestrated by a delicate interplay of biochemical and biophysical factors. While the role of genetics and biochemistry in embryogenesis has been extensively studied, recent research has highlighted the significance of mechanical regulation in shaping and guiding this intricate process. Here, we provide an overview of the current understanding of the mechanical regulation of embryo development. We explore how mechanical forces generated by cells and tissues play a crucial role in driving the development of different stages. We examine key morphogenetic processes such as compaction, blastocyst formation, implantation, and egg cylinder formation, and discuss the mechanical mechanisms and cues involved. By synthesizing the current body of literature, we highlight the emerging concepts and open questions in the field of mechanical regulation. We aim to provide an overview of the field, inspiring future investigations and fostering a deeper understanding of the mechanical aspects of embryo development.
Collapse
Affiliation(s)
- Hong Huang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China;
| | - Min Bao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China;
| |
Collapse
|
9
|
Geisler HC, Safford HC, Mitchell MJ. Rational Design of Nanomedicine for Placental Disorders: Birthing a New Era in Women's Reproductive Health. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300852. [PMID: 37191231 PMCID: PMC10651803 DOI: 10.1002/smll.202300852] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/16/2023] [Indexed: 05/17/2023]
Abstract
The placenta is a transient organ that forms during pregnancy and acts as a biological barrier, mediating exchange between maternal and fetal circulation. Placental disorders, such as preeclampsia, fetal growth restriction, placenta accreta spectrum, and gestational trophoblastic disease, originate in dysfunctional placental development during pregnancy and can lead to severe complications for both the mother and fetus. Unfortunately, treatment options for these disorders are severely lacking. Challenges in designing therapeutics for use during pregnancy involve selectively delivering payloads to the placenta while protecting the fetus from potential toxic side effects. Nanomedicine holds great promise in overcoming these barriers; the versatile and modular nature of nanocarriers, including prolonged circulation times, intracellular delivery, and organ-specific targeting, can control how therapeutics interact with the placenta. In this review, nanomedicine strategies are discussed to treat and diagnose placental disorders with an emphasis on understanding the unique pathophysiology behind each of these diseases. Finally, prior study of the pathophysiologic mechanisms underlying these placental disorders has revealed novel disease targets. These targets are highlighted here to motivate the rational design of precision nanocarriers to improve therapeutic options for placental disorders.
Collapse
Affiliation(s)
- Hannah C. Geisler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hannah C. Safford
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19014, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| |
Collapse
|
10
|
Lintao RCV, Richardson LS, Kammala AK, Chapa J, Yunque-Yap DA, Khanipov K, Golovko G, Dalmacio LMM, Menon R. PGRMC2 and HLA-G regulate immune homeostasis in a microphysiological model of human maternal-fetal membrane interface. Commun Biol 2024; 7:1041. [PMID: 39179795 PMCID: PMC11344061 DOI: 10.1038/s42003-024-06740-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024] Open
Abstract
Chorion trophoblasts (CTCs) and immune cell-enriched decidua (DECs) comprise the maternal-fetal membrane interface called the chorio-decidual interface (CDi) which constantly gets exposed to maternal stressors without leading to labor activation. This study explored how CTCs act as a barrier at CDi. The roles of human leukocyte antigen (HLA)-G and progesterone receptor membrane component 2 (PGRMC2) in mediating immune homeostasis were also investigated. The CDi was recreated in a two-chamber microfluidic device (CDi-on-chip) with an outer chamber of primary DECs and immune cell line-derived innate immune cells and an inner chamber of wild-type or PGRMC2 or HLA-G knockout immortalized CTCs. To mimic maternal insults, DECs were treated with lipopolysaccharide, poly(I:C), or oxidative stress inducer cigarette smoke extract. Expression levels of inflammation and immunity genes via targeted RNA sequencing, production of soluble mediators, and immune cell migration into CTCs were determined. In CDi-on-chip, decidua and immune cells became inflammatory in response to insults while CTCs were refractory, highlighting their barrier function. HLA-G and PGRMC2 are found to be vital to immune homeostasis at the CDi, with PGRMC2 serving as an upstream regulator of inflammation, HLA-G expression, and mesenchymal-epithelial transition, and HLA-G serving as a frontline immunomodulatory molecule, thus preventing fetal membrane compromise.
Collapse
Affiliation(s)
- Ryan C V Lintao
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
- Institute of Reproductive Health, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
| | - Lauren S Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Ananth Kumar Kammala
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Jenieve Chapa
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Dianne Aster Yunque-Yap
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Microbiome and Bioinformatics Analysis Core, The Institute for Translational Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- National Aeronautics and Space Administration Johnson Space Center, Houston, TX, USA
- KBR, Houston, TX, USA
| | - Kamil Khanipov
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Microbiome and Bioinformatics Analysis Core, The Institute for Translational Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - George Golovko
- Department of Pharmacology and Toxicology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
- Microbiome and Bioinformatics Analysis Core, The Institute for Translational Sciences, The University of Texas Medical Branch at Galveston, Galveston, TX, USA
| | - Leslie Michelle M Dalmacio
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Manila, Philippines
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, USA.
| |
Collapse
|
11
|
Lopez I, Truskey GA. Multi-cellular engineered living systems to assess reproductive toxicology. Reprod Toxicol 2024; 127:108609. [PMID: 38759876 PMCID: PMC11179964 DOI: 10.1016/j.reprotox.2024.108609] [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: 12/21/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/19/2024]
Abstract
Toxicants and some drugs can negatively impact reproductive health. Many toxicants haven't been tested due to lack of available models. The impact of many drugs taken during pregnancy to address maternal health may adversely affect fetal development with life-long effects and clinical trials do not examine toxicity effects on the maternal-fetal interface, requiring indirect assessment of safety and efficacy. Due to current gaps in reproductive toxicological knowledge and limitations of animal models, multi-cellular engineered living systems may provide solutions for modeling reproductive physiology and pathology for chemical and xenobiotic toxicity studies. Multi-cellular engineered living systems, such as microphysiological systems (MPS) and organoids, model of functional units of tissues. In this review, we highlight the key functions and structures of human reproductive organs and well-known representative toxicants afflicting these systems. We then discuss current approaches and specific studies where scientists have used MPS or organoids to recreate in vivo markers and cellular responses of the female and male reproductive system, as well as pregnancy-associated placenta formation and embryo development. We provide specific examples of organoids and organ-on-chip that have been used for toxicological purposes with varied success. Finally, we address current issues related to usage of MPS, emerging techniques for improving upon these complications, and improvements needed to make MPS more capable in assessing reproductive toxicology. Overall, multi-cellular engineered living systems have considerable promise to serve as a suitable, alternative reproductive biological model compared to animal studies and 2D culture.
Collapse
Affiliation(s)
- Isabella Lopez
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States.
| |
Collapse
|
12
|
Park S, Hunter ES. Modeling the human placenta: in vitro applications in developmental and reproductive toxicology. Crit Rev Toxicol 2024; 54:431-464. [PMID: 39016688 DOI: 10.1080/10408444.2023.2295349] [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: 07/31/2023] [Revised: 11/20/2023] [Accepted: 12/07/2023] [Indexed: 07/18/2024]
Abstract
During its temporary tenure, the placenta has extensive and specialized functions that are critical for pre- and post-natal development. The consequences of chemical exposure in utero can have profound effects on the structure and function of pregnancy-associated tissues and the life-long health of the birthing person and their offspring. However, the toxicological importance and critical functions of the placenta to embryonic and fetal development and maturation have been understudied. This narrative will review early placental development in humans and highlight some in vitro models currently in use that are or can be applied to better understand placental processes underlying developmental toxicity due to in utero environmental exposures.
Collapse
Affiliation(s)
- Sarah Park
- Oak Ridge Institute for Science and Education (ORISE), Oak Ridge, TN, USA
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| | - Edward Sidney Hunter
- Center for Computational Toxicology and Exposure, ORD, US EPA, Research Triangle Park, NC, USA
| |
Collapse
|
13
|
Wu T, Yan J, Nie K, Chen Y, Wu Y, Wang S, Zhang J. Microfluidic chips in female reproduction: a systematic review of status, advances, and challenges. Theranostics 2024; 14:4352-4374. [PMID: 39113805 PMCID: PMC11303079 DOI: 10.7150/thno.97301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/06/2024] [Indexed: 08/10/2024] Open
Abstract
The female reproductive system is essential to women's health, human reproduction and societal well-being. However, the clinical translation of traditional research models is restricted due to the uncertain effects and low efficiency. Emerging evidence shows that microfluidic chips provide valuable platforms for studying the female reproductive system, while no paper has ever comprehensively discussed the topic. Here, a total of 161 studies out of 14,669 records are identified in PubMed, Scopus, Web of Science, ScienceDirect and IEEE Xplore databases. Among these, 61 studies focus on oocytes, which further involves culture, cell surgeries (oocyte separation, rotation, enucleation, and denudation), evaluation and cryopreservation. Forty studies investigate embryo manipulation via microfluidic chips, covering in vitro fertilization, cryopreservation and functional evaluation. Forty-six studies reconstitute both the physiological and pathological statuses of in vivo organs, mostly involved in placenta and fetal membrane research. Fourteen studies perform drug screening and toxicity testing. In this review, we summarize the current application of microfluidic chips in studying the female reproductive system, the advancements in materials and methods, and discuss the future challenges. The present evidence suggests that microfluidic chips-assisted reproductive system reconstruction is promising and more studies are urgently needed.
Collapse
Affiliation(s)
- Tong Wu
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinfeng Yan
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, China
| | - Kebing Nie
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangyang Wu
- College of Animal Science and Technology, Sichuan Agricultural University, Sichuan, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
14
|
Horvat Mercnik M, Schliefsteiner C, Sanchez-Duffhues G, Wadsack C. TGFβ signalling: a nexus between inflammation, placental health and preeclampsia throughout pregnancy. Hum Reprod Update 2024; 30:442-471. [PMID: 38519450 PMCID: PMC11215164 DOI: 10.1093/humupd/dmae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/16/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The placenta is a unique and pivotal organ in reproduction, controlling crucial growth and cell differentiation processes that ensure a successful pregnancy. Placental development is a tightly regulated and dynamic process, in which the transforming growth factor beta (TGFβ) superfamily plays a central role. This family of pleiotropic growth factors is heavily involved in regulating various aspects of reproductive biology, particularly in trophoblast differentiation during the first trimester of pregnancy. TGFβ signalling precisely regulates trophoblast invasion and the cell transition from cytotrophoblasts to extravillous trophoblasts, which is an epithelial-to-mesenchymal transition-like process. Later in pregnancy, TGFβ signalling ensures proper vascularization and angiogenesis in placental endothelial cells. Beyond its role in trophoblasts and endothelial cells, TGFβ signalling contributes to the polarization and function of placental and decidual macrophages by promoting maternal tolerance of the semi-allogeneic foetus. Disturbances in early placental development have been associated with several pregnancy complications, including preeclampsia (PE) which is one of the severe complications. Emerging evidence suggests that TGFβ is involved in the pathogenesis of PE, thereby offering a potential target for intervention in the human placenta. OBJECTIVE AND RATIONALE This comprehensive review aims to explore and elucidate the roles of the major members of the TGFβ superfamily, including TGFβs, bone morphogenetic proteins (BMPs), activins, inhibins, nodals, and growth differentiation factors (GDFs), in the context of placental development and function. The review focusses on their interactions within the major cell types of the placenta, namely trophoblasts, endothelial cells, and immune cells, in both normal pregnancies and pregnancies complicated by PE throughout pregnancy. SEARCH METHODS A literature search was carried out using PubMed and Google Scholar, searching terms: 'TGF signalling preeclampsia', 'pregnancy TGF signalling', 'preeclampsia tgfβ', 'preeclampsia bmp', 'preeclampsia gdf', 'preeclampsia activin', 'endoglin preeclampsia', 'endoglin pregnancy', 'tgfβ signalling pregnancy', 'bmp signalling pregnancy', 'gdf signalling pregnancy', 'activin signalling pregnancy', 'Hofbauer cell tgfβ signalling', 'placental macrophages tgfβ', 'endothelial cells tgfβ', 'endothelium tgfβ signalling', 'trophoblast invasion tgfβ signalling', 'trophoblast invasion Smad', 'trophoblast invasion bmp', 'trophoblast invasion tgfβ', 'tgfβ preeclampsia', 'tgfβ placental development', 'TGFβ placental function', 'endothelial dysfunction preeclampsia tgfβ signalling', 'vascular remodelling placenta TGFβ', 'inflammation pregnancy tgfβ', 'immune response pregnancy tgfβ', 'immune tolerance pregnancy tgfβ', 'TGFβ pregnancy NK cells', 'bmp pregnancy NK cells', 'bmp pregnancy tregs', 'tgfβ pregnancy tregs', 'TGFβ placenta NK cells', 'TGFβ placenta tregs', 'NK cells preeclampsia', 'Tregs preeclampsia'. Only articles published in English until 2023 were used. OUTCOMES A comprehensive understanding of TGFβ signalling and its role in regulating interconnected cell functions of the main placental cell types provides valuable insights into the processes essential for successful placental development and growth of the foetus during pregnancy. By orchestrating trophoblast invasion, vascularization, immune tolerance, and tissue remodelling, TGFβ ligands contribute to the proper functioning of a healthy maternal-foetal interface. However, dysregulation of TGFβ signalling has been implicated in the pathogenesis of PE, where the shallow trophoblast invasion, defective vascular remodelling, decreased uteroplacental perfusion, and endothelial cell and immune dysfunction observed in PE, are all affected by an altered TGFβ signalling. WIDER IMPLICATIONS The dysregulation of TGFβ signalling in PE has important implications for research and clinical practice. Further investigation is required to understand the underlying mechanisms, including the role of different ligands and their regulation under pathophysiological conditions, in order to discover new therapeutic targets. Distinguishing between clinically manifested subtypes of PE and studying TGFβ signalling in different placental cell types holistically is an important first step. To put this knowledge into practice, pre-clinical animal models combined with new technologies are needed. This may also lead to improved human research models and identify potential therapeutic targets, ultimately improving outcomes for affected pregnancies and reducing the burden of PE.
Collapse
Affiliation(s)
| | | | - Gonzalo Sanchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Tissue-Specific BMP Signalling ISPA-HUCA, Oviedo, Spain
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
15
|
Nwokoye PN, Abilez OJ. Bioengineering methods for vascularizing organoids. CELL REPORTS METHODS 2024; 4:100779. [PMID: 38759654 PMCID: PMC11228284 DOI: 10.1016/j.crmeth.2024.100779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 03/01/2024] [Accepted: 04/24/2024] [Indexed: 05/19/2024]
Abstract
Organoids, self-organizing three-dimensional (3D) structures derived from stem cells, offer unique advantages for studying organ development, modeling diseases, and screening potential therapeutics. However, their translational potential and ability to mimic complex in vivo functions are often hindered by the lack of an integrated vascular network. To address this critical limitation, bioengineering strategies are rapidly advancing to enable efficient vascularization of organoids. These methods encompass co-culturing organoids with various vascular cell types, co-culturing lineage-specific organoids with vascular organoids, co-differentiating stem cells into organ-specific and vascular lineages, using organoid-on-a-chip technology to integrate perfusable vasculature within organoids, and using 3D bioprinting to also create perfusable organoids. This review explores the field of organoid vascularization, examining the biological principles that inform bioengineering approaches. Additionally, this review envisions how the converging disciplines of stem cell biology, biomaterials, and advanced fabrication technologies will propel the creation of increasingly sophisticated organoid models, ultimately accelerating biomedical discoveries and innovations.
Collapse
Affiliation(s)
- Peter N Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Oscar J Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA; Division of Pediatric CT Surgery, Stanford University, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; Maternal and Child Health Research Institute, Stanford University, Stanford, CA 94305, USA; Bio-X Program, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
16
|
Nwokoye PN, Abilez OJ. Blood vessels in a dish: the evolution, challenges, and potential of vascularized tissues and organoids. Front Cardiovasc Med 2024; 11:1336910. [PMID: 38938652 PMCID: PMC11210405 DOI: 10.3389/fcvm.2024.1336910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/19/2024] [Indexed: 06/29/2024] Open
Abstract
Vascular pathologies are prevalent in a broad spectrum of diseases, necessitating a deeper understanding of vascular biology, particularly in overcoming the oxygen and nutrient diffusion limit in tissue constructs. The evolution of vascularized tissues signifies a convergence of multiple scientific disciplines, encompassing the differentiation of human pluripotent stem cells (hPSCs) into vascular cells, the development of advanced three-dimensional (3D) bioprinting techniques, and the refinement of bioinks. These technologies are instrumental in creating intricate vascular networks essential for tissue viability, especially in thick, complex constructs. This review provides broad perspectives on the past, current state, and advancements in key areas, including the differentiation of hPSCs into specific vascular lineages, the potential and challenges of 3D bioprinting methods, and the role of innovative bioinks mimicking the native extracellular matrix. We also explore the integration of biophysical cues in vascularized tissues in vitro, highlighting their importance in stimulating vessel maturation and functionality. In this review, we aim to synthesize these diverse yet interconnected domains, offering a broad, multidisciplinary perspective on tissue vascularization. Advancements in this field will help address the global organ shortage and transform patient care.
Collapse
Affiliation(s)
- Peter N. Nwokoye
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Oscar J. Abilez
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, United States
- Division of Pediatric CT Surgery, Stanford University, Stanford, CA, United States
- Cardiovascular Institute, Stanford University, Stanford, CA, United States
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, United States
- Bio-X Program, Stanford University, Stanford, CA, United States
| |
Collapse
|
17
|
Lestari B, Fukushima T, Utomo RY, Wahyuningsih MSH. Apoptotic and non-apoptotic roles of caspases in placenta physiology and pathology. Placenta 2024; 151:37-47. [PMID: 38703713 DOI: 10.1016/j.placenta.2024.03.013] [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: 11/15/2023] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 05/06/2024]
Abstract
Caspases, a family of cysteine proteases, are pivotal regulators of apoptosis, the tightly controlled cell death process crucial for eliminating excessive or unnecessary cells during development, including placental development. Collecting research has unveiled the multifaceted roles of caspases in the placenta, extending beyond apoptosis. Apart from their involvement in placental tissue remodeling via apoptosis, caspases actively participate in essential regulatory processes, such as trophoblast fusion and differentiation, significantly influencing placental growth and functionality. In addition, growing evidence indicates an elevation in caspase activity under pathological conditions like pre-eclampsia (PE) and intrauterine growth restriction (IUGR), leading to excessive cell death as well as inflammation. Drawing from advancements in caspase research and placental development under both normal and abnormal conditions, we examine the significance of caspases in both cell death (apoptosis) and non-cell death-related processes within the placenta. We also discuss potential therapeutics targeting caspase-related pathways for placenta disorders.
Collapse
Affiliation(s)
- Beni Lestari
- Department Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia; Cancer Chemoprevention Research Center, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Toshiaki Fukushima
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Japan.
| | - Rohmad Yudi Utomo
- Cancer Chemoprevention Research Center, Universitas Gadjah Mada, Yogyakarta, Indonesia; Department Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Mae Sri Hartati Wahyuningsih
- Department Pharmacology and Therapy, Faculty of Medicine, Public Health and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia.
| |
Collapse
|
18
|
Golden TN, Mani S, Linn RL, Leite R, Trigg NA, Wilson A, Anton L, Mainigi M, Conine CC, Kaufman BA, Strauss JF, Parry S, Simmons RA. Extracellular vesicles alter trophoblast function in pregnancies complicated by COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.17.580824. [PMID: 38464046 PMCID: PMC10925147 DOI: 10.1101/2024.02.17.580824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and resulting coronavirus disease (COVID-19) causes placental dysfunction, which increases the risk of adverse pregnancy outcomes. While abnormal placental pathology resulting from COVID-19 is common, direct infection of the placenta is rare. This suggests that pathophysiology associated with maternal COVID-19, rather than direct placental infection, is responsible for placental dysfunction and alteration of the placental transcriptome. We hypothesized that maternal circulating extracellular vesicles (EVs), altered by COVID-19 during pregnancy, contribute to placental dysfunction. To examine this hypothesis, we characterized maternal circulating EVs from pregnancies complicated by COVID-19 and tested their effects on trophoblast cell physiology in vitro . We found that the gestational timing of COVID-19 is a major determinant of circulating EV function and cargo. In vitro trophoblast exposure to EVs isolated from patients with an active infection at the time of delivery, but not EVs isolated from Controls, altered key trophoblast functions including hormone production and invasion. Thus, circulating EVs from participants with an active infection, both symptomatic and asymptomatic cases, can disrupt vital trophoblast functions. EV cargo differed between participants with COVID-19 and Controls, which may contribute to the disruption of the placental transcriptome and morphology. Our findings show that COVID-19 can have effects throughout pregnancy on circulating EVs and circulating EVs are likely to participate in placental dysfunction induced by COVID-19.
Collapse
|
19
|
Jeong S, Fuwad A, Yoon S, Jeon TJ, Kim SM. A Microphysiological Model to Mimic the Placental Remodeling during Early Stage of Pregnancy under Hypoxia-Induced Trophoblast Invasion. Biomimetics (Basel) 2024; 9:289. [PMID: 38786499 PMCID: PMC11118815 DOI: 10.3390/biomimetics9050289] [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: 03/18/2024] [Revised: 05/05/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Placental trophoblast invasion is critical for establishing the maternal-fetal interface, yet the mechanisms driving trophoblast-induced maternal arterial remodeling remain elusive. To address this gap, we developed a three-dimensional microfluidic placenta-on-chip model that mimics early pregnancy placentation in a hypoxic environment. By studying human umbilical vein endothelial cells (HUVECs) under oxygen-deprived conditions upon trophoblast invasion, we observed significant HUVEC artery remodeling, suggesting the critical role of hypoxia in placentation. In particular, we found that trophoblasts secrete matrix metalloproteinase (MMP) proteins under hypoxic conditions, which contribute to arterial remodeling by the degradation of extracellular matrix components. This MMP-mediated remodeling is critical for facilitating trophoblast invasion and proper establishment of the maternal-fetal interface. In addition, our platform allows real-time monitoring of HUVEC vessel contraction during trophoblast interaction, providing valuable insights into the dynamic interplay between trophoblasts and maternal vasculature. Collectively, our findings highlight the importance of MMP-mediated arterial remodeling in placental development and underscore the potential of our platform to study pregnancy-related complications and evaluate therapeutic interventions.
Collapse
Affiliation(s)
- Seorin Jeong
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.J.); (A.F.)
| | - Ahmed Fuwad
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.J.); (A.F.)
- Department of Biomedical Engineering, School of Mechanical & Manufacturing Engineering (SMME), National University of Science and Technology (NUST), Islamabad 44000, Pakistan
| | - Sunhee Yoon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea;
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea;
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Department of Biological Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Sun Min Kim
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.J.); (A.F.)
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea;
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
20
|
Shelton SE. Vascular microphysiological systems. Curr Opin Hematol 2024; 31:155-161. [PMID: 38236999 DOI: 10.1097/moh.0000000000000802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2024]
Abstract
PURPOSE OF REVIEW This review summarizes innovations in vascular microphysiological systems (MPS) and discusses the themes that have emerged from recent works. RECENT FINDINGS Vascular MPS are increasing in complexity and ability to replicate tissue. Many labs use vascular MPS to study transport phenomena such as analyzing endothelial barrier function. Beyond vascular permeability, these models are also being used for pharmacological studies, including drug distribution and toxicity modeling. In part, these studies are made possible due to exciting advances in organ-specific models. Inflammatory processes have also been modeled by incorporating immune cells, with the ability to explore both cell migration and function. Finally, as methods for generating vascular MPS flourish, many researchers have turned their attention to incorporating flow to more closely recapitulate in vivo conditions. SUMMARY These models represent many different types of tissue and disease states. Some devices have relatively simple geometry and few cell types, while others use complex, multicompartmental microfluidics and integrate several cell types and origins. These 3D models enable us to observe model evolution in real time and perform a plethora of functional assays not possible using traditional cell culture methods.
Collapse
Affiliation(s)
- Sarah E Shelton
- Joint Department of Biomedical Engineering at the University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
21
|
Li Z, Yu D, Zhou C, Wang F, Lu K, Liu Y, Xu J, Xuan L, Wang X. Engineering vascularised organoid-on-a-chip: strategies, advances and future perspectives. BIOMATERIALS TRANSLATIONAL 2024; 5:21-32. [PMID: 39220668 PMCID: PMC11362354 DOI: 10.12336/biomatertransl.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 09/04/2024]
Abstract
In recent years, advances in microfabrication technology and tissue engineering have propelled the development of a novel drug screening and disease modelling platform known as organoid-on-a-chip. This platform integrates organoids and organ-on-a-chip technologies, emerging as a promising approach for in vitro modelling of human organ physiology. Organoid-on-a-chip devices leverage microfluidic systems to simulate the physiological microenvironment of specific organs, offering a more dynamic and flexible setting that can mimic a more comprehensive human biological context. However, the lack of functional vasculature has remained a significant challenge in this technology. Vascularisation is crucial for the long-term culture and in vitro modelling of organoids, holding important implications for drug development and personalised medical approaches. This review provides an overview of research progress in developing vascularised organoid-on-a-chip models, addressing methods for in vitro vascularisation and advancements in vascularised organoids. The aim is to serve as a reference for future endeavors in constructing fully functional vascularised organoid-on-a-chip platforms.
Collapse
Affiliation(s)
- Zhangjie Li
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Dingyuan Yu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chenyang Zhou
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Feifan Wang
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Kangyi Lu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yijun Liu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jiaqi Xu
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Lian Xuan
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolin Wang
- Department of Micro/Nano Electronics, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai, China
- National Key Laboratory of Advanced Micro and Nano Manufacture Technology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
22
|
Mani S, Garifallou J, Kim SJ, Simoni MK, Huh DD, Gordon SM, Mainigi M. Uterine macrophages and NK cells exhibit population and gene-level changes after implantation but maintain pro-invasive properties. Front Immunol 2024; 15:1364036. [PMID: 38566989 PMCID: PMC10985329 DOI: 10.3389/fimmu.2024.1364036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Introduction Prior to pregnancy, hormonal changes lead to cellular adaptations in the endometrium allowing for embryo implantation. Critical for successful pregnancy establishment, innate immune cells constitute a significant proportion of uterine cells prior to arrival of the embryo and throughout the first trimester in humans and animal models. Abnormal uterine immune cell function during implantation is believed to play a role in multiple adverse pregnancy outcomes. Current work in humans has focused on uterine immune cells present after pregnancy establishment, and limited in vitro models exist to explore unique functions of these cells. Methods With single-cell RNA-sequencing (scRNAseq), we comprehensively compared the human uterine immune landscape of the endometrium during the window of implantation and the decidua during the first trimester of pregnancy. Results We uncovered global and cell-type-specific gene signatures for each timepoint. Immune cells in the endometrium prior to implantation expressed genes associated with immune metabolism, division, and activation. In contrast, we observed widespread interferon signaling during the first trimester of pregnancy. We also provide evidence of specific inflammatory pathways enriched in pre- and post-implantation macrophages and natural killer (NK) cells in the uterine lining. Using our novel implantation-on-a-chip (IOC) to model human implantation ex vivo, we demonstrate for the first time that uterine macrophages strongly promote invasion of extravillous trophoblasts (EVTs), a process essential for pregnancy establishment. Pre- and post-implantation uterine macrophages promoted EVT invasion to a similar degree as pre- and post-implantation NK cells on the IOC. Conclusions This work provides a foundation for further investigation of the individual roles of uterine immune cell subtypes present prior to embryo implantation and during early pregnancy, which will be critical for our understanding of pregnancy complications associated with abnormal trophoblast invasion and placentation.
Collapse
Affiliation(s)
- Sneha Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - James Garifallou
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Se-jeong Kim
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Michael K. Simoni
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Dan Dongeun Huh
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States
- National Science Foundation (NSF) Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Scott M. Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| | - Monica Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
23
|
Simoni MK, Negatu SG, Park JY, Mani S, Arreguin MC, Amses K, Huh DD, Mainigi M, Jurado KA. Type I interferon alters invasive extravillous trophoblast function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584521. [PMID: 38559122 PMCID: PMC10979977 DOI: 10.1101/2024.03.11.584521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Inappropriate type I interferon (IFN) signaling during embryo implantation and placentation is linked to poor pregnancy outcomes. Here, we evaluated the consequence of elevated type I IFN exposure on implantation using a biomimetic model of human implantation in an organ-on-a-chip device. We found that type I IFN reduced extravillous trophoblast (EVT) invasion capacity. Analyzing single-cell transcriptomes, we uncovered that IFN truncated endovascular EVT emergence in the implantation-on-a-chip device by stunting EVT epithelial-to-mesenchymal transition. Disruptions to the epithelial-to-mesenchymal transition is associated with the pathogenesis of preeclampsia, a life-threatening hypertensive disorder of pregnancy. Strikingly, unwarranted IFN stimulation induced genes associated with increased preeclampsia risk and a preeclamptic gene-like signature in EVTs. These dysregulated EVT phenotypes ultimately reduced EVT-mediated endothelial cell vascular remodeling in the implantation-on-a-chip device. Overall, our work indicates IFN signaling can alter EVT epithelial-to-mesenchymal transition progression which results in diminished EVT-mediated spiral artery remodeling and a preeclampsia gene signature upon sustained stimulation. Our work implicates unwarranted type I IFN as a maternal disturbance that can result in abnormal EVT function that could trigger preeclampsia.
Collapse
|
24
|
Xu Y, Liu X, Zeng W, Zhu Y, Dong J, Wu F, Chen C, Sharma S, Lin Y. DOCK1 insufficiency disrupts trophoblast function and pregnancy outcomes via DUSP4-ERK pathway. Life Sci Alliance 2024; 7:e202302247. [PMID: 37967942 PMCID: PMC10651491 DOI: 10.26508/lsa.202302247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023] Open
Abstract
Abnormal trophoblast function is associated with diseases such as recurrent spontaneous abortion, pre-eclampsia, and preterm birth, and endangers maternal and fetal health. However, the underlying regulatory mechanisms remain unclear. In this study, we found DOCK1 expression is decreased in the placental villi of patients with recurrent spontaneous abortion, and that its expression determined the invasive properties of extravillous trophoblasts (EVTs), highlighting a previously unknown role of DOCK1 in regulating EVT function. Furthermore, DOCK1 deficiency disturbed the ubiquitinated degradation of DUSP4, leading to its accumulation. This caused inactivation of the ERK signaling pathway, resulting in inadequate EVT migration and invasion. DOCK1 was implicated in regulating the ubiquitin levels of DUSP4, possibly by modulating the E3 ligase enzyme HUWE1. The results of our in vivo experiments confirmed that the DOCK1 inhibitor TBOPP caused miscarriage in mice by inactivating the DUSP4/ERK pathway. Collectively, our results revealed the crucial role of DOCK1 in the regulation of EVT function via the DUSP4-ERK pathway and a basis for the development of novel treatments for adverse pregnancy outcomes caused by trophoblast dysfunction.
Collapse
Affiliation(s)
- Yichi Xu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaorui Liu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weihong Zeng
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yueyue Zhu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Junpeng Dong
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Wu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Institute of Birth Defects and Rare Diseases, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cailian Chen
- Department of Automation, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, China
- Key Laboratory of System Control and Information Processing, Ministry of Education of China, Shanghai, China
| | - Surendra Sharma
- Department of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Yi Lin
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
25
|
Li Q, Sharkey A, Sheridan M, Magistrati E, Arutyunyan A, Huhn O, Sancho-Serra C, Anderson H, McGovern N, Esposito L, Fernando R, Gardner L, Vento-Tormo R, Turco MY, Moffett A. Human uterine natural killer cells regulate differentiation of extravillous trophoblast early in pregnancy. Cell Stem Cell 2024; 31:181-195.e9. [PMID: 38237587 DOI: 10.1016/j.stem.2023.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 10/19/2023] [Accepted: 12/20/2023] [Indexed: 02/04/2024]
Abstract
In humans, balanced invasion of trophoblast cells into the uterine mucosa, the decidua, is critical for successful pregnancy. Evidence suggests that this process is regulated by uterine natural killer (uNK) cells, but how they influence reproductive outcomes is unclear. Here, we used our trophoblast organoids and primary tissue samples to determine how uNK cells affect placentation. By locating potential interaction axes between trophoblast and uNK cells using single-cell transcriptomics and in vitro modeling of these interactions in organoids, we identify a uNK cell-derived cytokine signal that promotes trophoblast differentiation at the late stage of the invasive pathway. Moreover, it affects transcriptional programs involved in regulating blood flow, nutrients, and inflammatory and adaptive immune responses, as well as gene signatures associated with disorders of pregnancy such as pre-eclampsia. Our findings suggest mechanisms on how optimal immunological interactions between uNK cells and trophoblast enhance reproductive success.
Collapse
Affiliation(s)
- Qian Li
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| | - Andrew Sharkey
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Megan Sheridan
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Elisa Magistrati
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Anna Arutyunyan
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Oisin Huhn
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Carmen Sancho-Serra
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Holly Anderson
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Naomi McGovern
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Laura Esposito
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Ridma Fernando
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Lucy Gardner
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Roser Vento-Tormo
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK; Wellcome Sanger Institute, Cambridge CB10 1SA, UK.
| | | | - Ashley Moffett
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK; Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK.
| |
Collapse
|
26
|
Li D, Jie Q, Li Q, Long P, Wang Z, Wang J, Tian S, Wu M, Ma Y, Huang Y. CsA promotes trophoblast invasion accompanied by changes in leukaemic inhibitory factor and fibroblast growth factor in peri-implantation blastocysts. ZYGOTE 2024; 32:71-76. [PMID: 38124629 DOI: 10.1017/s0967199423000497] [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: 12/23/2023]
Abstract
During the early stages of human pregnancy, successful implantation of embryonic trophoblast cells into the endometrium depends on good communication between trophoblast cells and the endometrium. Abnormal trophoblast cell function can cause embryo implantation failure. In this study, we added cyclosporine A (CsA) to the culture medium to observe the effect of CsA on embryonic trophoblast cells and the related mechanism. We observed that CsA promoted the migration and invasion of embryonic trophoblast cells. CsA promoted the expression of leukaemic inhibitory factor (LIF) and fibroblast growth factor (FGF). In addition, CsA promoted the secretion and volume increase in vesicles in the CsA-treated group compared with the control group. Therefore, CsA may promote the adhesion and invasion of trophoblast cells through LIF and FGF and promote the vesicle dynamic process, which is conducive to embryo implantation.
Collapse
Affiliation(s)
- Dan Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Hainan Medical University, China
- Department of Reproductive Medicine, Haikou Women & Children Hospital, China
| | - Qiuling Jie
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Hainan Medical University, China
- Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
| | - Qi Li
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
| | - Ping Long
- Guizhou Qiannan People's Hospital, China
| | - Zhen Wang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
| | | | | | - Menglan Wu
- Department of Reproductive Medicine, The First Affiliated Hospital of Zhengzhou University, China
| | - Yanlin Ma
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Hainan Medical University, China
- Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
| | - Yuanhua Huang
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Hainan Medical University, China
- Hainan Provincial Clinical Research Center for Thalassemia, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Department of Reproductive Medicine, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
- Haikou Key Laboratory for Preservation of Human Genetic Resource, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, China
| |
Collapse
|
27
|
Yan J, Wu T, Zhang J, Gao Y, Wu JM, Wang S. Revolutionizing the female reproductive system research using microfluidic chip platform. J Nanobiotechnology 2023; 21:490. [PMID: 38111049 PMCID: PMC10729361 DOI: 10.1186/s12951-023-02258-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
Comprehensively understanding the female reproductive system is crucial for safeguarding fertility and preventing diseases concerning women's health. With the capacity to simulate the intricate physio- and patho-conditions, and provide diagnostic platforms, microfluidic chips have fundamentally transformed the knowledge and management of female reproductive health, which will ultimately promote the development of more effective assisted reproductive technologies, treatments, and drug screening approaches. This review elucidates diverse microfluidic systems in mimicking the ovary, fallopian tube, uterus, placenta and cervix, and we delve into the culture of follicles and oocytes, gametes' manipulation, cryopreservation, and permeability especially. We investigate the role of microfluidics in endometriosis and hysteromyoma, and explore their applications in ovarian cancer, endometrial cancer and cervical cancer. At last, the current status of assisted reproductive technology and integrated microfluidic devices are introduced briefly. Through delineating the multifarious advantages and challenges of the microfluidic technology, we chart a definitive course for future research in the woman health field. As the microfluidic technology continues to evolve and advance, it holds great promise for revolutionizing the diagnosis and treatment of female reproductive health issues, thus propelling us into a future where we can ultimately optimize the overall wellbeing and health of women everywhere.
Collapse
Affiliation(s)
- Jinfeng Yan
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Tong Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jinjin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Yueyue Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jia-Min Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
28
|
Mihalic ZN, Kloimböck T, Cosic-Mujkanovic N, Valadez-Cosmes P, Maitz K, Kindler O, Wadsack C, Heinemann A, Marsche G, Gauster M, Pollheimer J, Kargl J. Myeloperoxidase enhances the migration and invasion of human choriocarcinoma JEG-3 cells. Redox Biol 2023; 67:102885. [PMID: 37776707 PMCID: PMC10556814 DOI: 10.1016/j.redox.2023.102885] [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: 04/13/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
Myeloperoxidase (MPO) is one of the most abundant proteins in neutrophil granules. It catalyzes the production of reactive oxygen species, which are important in inflammation and immune defense. MPO also binds to several proteins, lipids, and DNA to alter their function. MPO is present at the feto-maternal interface during pregnancy, where neutrophils are abundant. In this study, we determined the effect of MPO on JEG-3 human choriocarcinoma cells as a model of extravillous trophoblasts (EVTs) during early pregnancy. We found that MPO was internalized by JEG-3 cells and localized to the cytoplasm and nuclei. MPO internalization and activity enhanced JEG-3 cell migration and invasion, whereas this effect was impaired by pre-treating cells with heparin, to block cellular uptake, and MPO-activity inhibitor 4-ABAH. This study identifies a novel mechanism for the effect of MPO on EVT function during normal pregnancy and suggests a potential role of MPO in abnormal pregnancies.
Collapse
Affiliation(s)
- Z N Mihalic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - T Kloimböck
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - N Cosic-Mujkanovic
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - P Valadez-Cosmes
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - K Maitz
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - O Kindler
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria
| | - C Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - A Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - G Marsche
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria
| | - M Gauster
- Division of Cell Biology, Histology and Embryology, Medical University of Graz, Austria
| | - J Pollheimer
- Department of Obstetrics and Gynecology, Reproductive Biology Unit, Maternal-Fetal Immunology Group, Medical University of Vienna, Austria
| | - J Kargl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria; BioTechMed-Graz, 8010, Graz, Austria.
| |
Collapse
|
29
|
Zeng YT, Liu WF, Zheng PS, Li S. GDF15 deficiency hinders human trophoblast invasion to mediate pregnancy loss through downregulating Smad1/5 phosphorylation. iScience 2023; 26:107902. [PMID: 37766993 PMCID: PMC10520888 DOI: 10.1016/j.isci.2023.107902] [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: 01/30/2023] [Revised: 05/04/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Growth differentiation factor 15 (GDF15) belongs to the Transforming growth factor β(TGF-β) superfamily. The decrease of GDF15 in the serum of pregnant women was associated with miscarriage. Both IHC and ELISA assays showed that GDF15 in trophoblast tissue and serum of pregnant women who miscarried was significantly lower than in those who had a live birth. GDF15 deficiency was associated with embryo resorption in GDF15 knockout mice through CRIPSR editing. In addition, the migration and invasion ability of HTR-8/SVneo and JEG-3 cells were promoted by GDF15. Mechanistically, GDF15 increased Smad1/5 phosphorylation, resulting in upregulating SNAI1/2, VIMENTIN and downregulating E-CADHERIN. A dual-luciferase reporter assay confirmed that Smad-binding elements (SBE) and/or GC-rich motifs were activated and target genes such as SNAI1/2, SERPINE1, and TIMP3 were transcriptionally regulated by GDF15/Smad5 signaling. Therefore, our data revealed a crucial role of GDF15 on invasion of trophoblast by upregulating the activity of TGF-β/Smad1/5 pathway.
Collapse
Affiliation(s)
- Yu-Ting Zeng
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Wen-Fang Liu
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Peng-Sheng Zheng
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of the People’s Republic of China, Xi’an, Shaanxi, China
| | - Shan Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
30
|
Gao Z, Guo J, Gou B, Gu Z, Jia T, Ma S, Jiang L, Liu W, Zhou L, Gu Q. Microcarriers promote the through interface movement of mouse trophoblast stem cells by regulating stiffness. Bioact Mater 2023; 28:196-205. [PMID: 37250864 PMCID: PMC10220236 DOI: 10.1016/j.bioactmat.2023.05.007] [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: 01/19/2023] [Revised: 04/20/2023] [Accepted: 05/09/2023] [Indexed: 05/31/2023] Open
Abstract
Mechanical force is crucial in the whole process of embryonic development. However, the role of trophoblast mechanics during embryo implantation has rarely been studied. In this study, we constructed a model to explore the effect of stiffness changes in mouse trophoblast stem cells (mTSCs) on implantation: microcarrier was prepared by sodium alginate using a droplet microfluidics system, and mTSCs were attached to the microcarrier surface with laminin modifications, called T(micro). Compared with the spheroid, formed by the self-assembly of mTSCs (T(sph)), we could regulate the stiffness of the microcarrier, making the Young's modulus of mTSCs (367.70 ± 79.81 Pa) similar to that of the blastocyst trophoblast ectoderm (432.49 ± 151.90 Pa). Moreover, T(micro) contributes to improve the adhesion rate, expansion area and invasion depth of mTSCs. Further, T(micro) was highly expressed in tissue migration-related genes due to the activation of the Rho-associated coiled-coil containing protein kinase (ROCK) pathway at relatively similar modulus of trophoblast. Overall, our study explores the embryo implantation process with a new perspective, and provides theoretical support for understanding the effect of mechanics on embryo implantation.
Collapse
Affiliation(s)
- Zili Gao
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Jia Guo
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Bo Gou
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Zhen Gu
- Department of Chemistry and Biological Engineering, University of Science and Technology, Beijing, 100083, PR China
| | - Tan Jia
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Sinan Ma
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- School of Life Sciences, Northeast Agricultural University, Harbin, 150030, PR China
| | - Liyuan Jiang
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- School of Life Sciences, Northeast Agricultural University, Harbin, 150030, PR China
| | - Wenli Liu
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
| | - Lixun Zhou
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, PR China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, PR China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| |
Collapse
|
31
|
Sun S, Xue X, Fu J. Modeling development using microfluidics: bridging gaps to foster fundamental and translational research. Curr Opin Genet Dev 2023; 82:102097. [PMID: 37573835 PMCID: PMC11193336 DOI: 10.1016/j.gde.2023.102097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 07/01/2023] [Accepted: 07/16/2023] [Indexed: 08/15/2023]
Abstract
In vitro stem cell-derived embryo and organ models, termed embryoids and organoids, respectively, provide promising experimental tools to study physiological and pathological processes in mammalian development and organ formation. Most of current embryoid and organoid systems are developed using conventional three-dimensional cultures that lack controls of spatiotemporal extracellular signals. Microfluidics, an established technology for quantitative controls and quantifications of dynamic chemical and physical environments, has recently been utilized for developing next-generation embryoids and organoids in a controllable and reproducible manner. In this review, we summarize recent progress in constructing microfluidics-based embryoids and organoids. Development of these models demonstrates the successful applications of microfluidics in establishing morphogen gradients, accelerating medium transport, exerting mechanical forces, facilitating tissue coculture studies, and improving assay throughput, thus supporting using microfluidics for building next-generation embryoids and organoids for fundamental and translational research.
Collapse
Affiliation(s)
- Shiyu Sun
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xufeng Xue
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
32
|
Tian J, Yang J, Chen T, Yin Y, Li N, Li Y, Luo X, Dong E, Tan H, Ma Y, Li T. Generation of Human Endometrial Assembloids with a Luminal Epithelium using Air-Liquid Interface Culture Methods. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301868. [PMID: 37635169 PMCID: PMC10602567 DOI: 10.1002/advs.202301868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/30/2023] [Indexed: 08/29/2023]
Abstract
The endometrial lining of the uterus is essential for women's reproductive health and consists of several different types of epithelial and stromal cells. Although models such as gland-like structures (GLSs) and endometrial assembloids (EnAos) are successfully established, they lack an intact luminal epithelium, which makes it difficult to recapitulate endometrial receptivity. Here, a novel EnAo model (ALI-EnAo) is developed by combining endometrial epithelial cells (EnECs) and stromal cells (EnSCs) and using an improved matrix and air-liquid interface (ALI) culture method. ALI-EnAos exhibit intact EnSCs and glandular and luminal epithelia, which recapitulates human endometrium anatomy, cell composition, hormone-induced menstrual cycle changes, gene expression profiles, and dynamic ciliogenesis. The model suggests that EnSCs, together with the extracellular matrix and ALI culture conditions, contribute to EnAo phenotypes and characteristics reflective of the endometrial menstrual cycle. This enables to transcriptionally define endometrial cell subpopulations. It anticipates that ALI-EnAos will facilitate studies on embryo implantation, and endometrial growth, differentiation, and disease.
Collapse
Affiliation(s)
- Jiwen Tian
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Medical SchoolKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Jie Yang
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Tingwei Chen
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Yu Yin
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Nan Li
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Yunxiu Li
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Xingyu Luo
- Medical SchoolKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - E Dong
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Haoyang Tan
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Yanping Ma
- Department of Reproductive MedicineThe First People's Hospital of Yunnan ProvinceKunmingYunnan650021China
| | - Tianqing Li
- State Key Laboratory of Primate Biomedical ResearchInstitute of Primate Translational MedicineKunming University of Science and TechnologyKunmingYunnan650032China
- Yunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| |
Collapse
|
33
|
de Lima Castro M, Dos Passos RR, Justina VD, do Amaral WN, Giachini FR. Physiological and pathological evidence of O-GlcNAcylation regulation during pregnancy related process. Placenta 2023; 141:43-50. [PMID: 37210277 DOI: 10.1016/j.placenta.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
O-GlcNAcylation is a dynamic and reversible post-translational modification (PTM) controlled by the enzymes O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Changes in its expression lead to a breakdown in cellular homeostasis, which is linked to several pathological processes. Placentation and embryonic development are periods of high cell activity, and imbalances in cell signaling pathways can result in infertility, miscarriage, or pregnancy complications. O-GlcNAcylation is involved in cellular processes such as genome maintenance, epigenetic regulation, protein synthesis/degradation, metabolic pathways, signaling pathways, apoptosis, and stress response. Trophoblastic differentiation/invasion and placental vasculogenesis, as well as zygote viability and embryonic neuronal development, are all dependent on O-GlcNAcylation. This PTM is required for pluripotency, which is a required condition for embryonic development. Further, this pathway is a nutritional sensor and cell stress marker, which is primarily measured by the OGT enzyme and its product, protein O-GlcNAcylation. Yet, this post-translational modification is enrolled in metabolic and cardiovascular adaptations during pregnancy. Finally, evidence of how O-GlcNAc impacts pregnancy during pathological conditions such as hyperglycemia, gestational diabetes, hypertension, and stress disorders are reviewed. Considering this scenario, progress in understanding the role of O- GlcNAcylation in pregnancy is required.
Collapse
Affiliation(s)
- Marta de Lima Castro
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Vanessa Dela Justina
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil
| | - Waldemar Naves do Amaral
- Graduation Program in Health Sciences, Faculty of Medicine, Federal University of Goias, Goiânia, Brazil
| | - Fernanda Regina Giachini
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil; Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Brazil.
| |
Collapse
|
34
|
Mao J, Feng Y, Zheng Y, Gao Y, Zhang L, Sun X, Wu Y, Zhu X, Ma F. GPR65 inhibits human trophoblast cell adhesion through upregulation of MYLK and downregulation of fibronectin via cAMP-ERK signaling in a low pH environment. Cell Commun Signal 2023; 21:238. [PMID: 37723567 PMCID: PMC10506227 DOI: 10.1186/s12964-023-01249-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 07/28/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Extravillous trophoblasts (EVTs) are essential cells during the formation of the placenta, with the major function of invading the maternal decidua, anchoring the developing placenta to the uterus, remodeling uterine arteries, and regulating immune responses to prevent rejection. During early pregnancy, the decidua undergoes a hypoxic and acidic microenvironment, which has been shown to participate in tumor cell migration, invasion, growth, and angiogenesis. Nevertheless, the mechanisms by which EVTs sense and respond to the acidic microenvironment, thereby executing their functions, remain poorly understood. METHODS The effects of G protein-coupled receptor 65 (GPR65) on cell adhesion and other cellular functions were tested using JAR spheroids, mouse blastocysts, and HTR-8/SVneo cells. Specifically, we employed HTR-8/SVneo cells for gene overexpression and silencing to investigate the underlying mechanism of GPR65's impact on trophoblast cell function under acidic conditions. Additionally, villus tissue samples obtained from early pregnancy loss patients were utilized to explore the potential association between GPR65 and its related signaling pathway molecules with the disease. RESULTS This study identified GPR65 expression widely in trophoblasts, with the highest level in EVTs. Importantly, optimal GPR65 levels are required for maintaining normal adhesion, migration, and invasion, whereas overexpression of GPR65 inhibits these functions by activating the cAMP-ERK signaling pathway, upregulating myosin light chain kinase (MYLK) and MYLK3 expression, and subsequently downregulating fibronectin. Consistently, elevated expression of GPR65, MYLK, and MYLK3 is observed in patients suffering from early pregnancy loss. CONCLUSIONS This work offers insights into the suppressive effects of GPR65 on EVT function under acidic conditions and highlights a putative target for therapeutic intervention in early pregnancy complications. Video Abstract.
Collapse
Affiliation(s)
- Jia Mao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yayun Zheng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaqiu Gao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China
| | - Linyu Zhang
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xinrui Sun
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yilun Wu
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaofeng Zhu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China.
| | - Fang Ma
- Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
35
|
Ren Y, Zhu D, Han X, Zhang Q, Chen B, Zhou P, Wei Z, Zhang Z, Cao Y, Zou H. HMGB1: a double-edged sword and therapeutic target in the female reproductive system. Front Immunol 2023; 14:1238785. [PMID: 37691930 PMCID: PMC10484633 DOI: 10.3389/fimmu.2023.1238785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
HMGB1 that belongs to the High Mobility Group-box superfamily, is a nonhistone chromatin associated transcription factor. It is present in the nucleus of eukaryotes and can be actively secreted or passively released by kinds of cells. HMGB1 is important for maintaining DNA structure by binding to DNA and histones, protecting it from damage. It also regulates the interaction between histones and DNA, affecting chromatin packaging, and can influence gene expression by promoting nucleosome sliding. And as a DAMP, HMGB1 binding to RAGE and TLRs activates NF-κB, which triggers the expression of downstream genes like IL-18, IL-1β, and TNF-α. HMGB1 is known to be involved in numerous physiological and pathological processes. Recent studies have demonstrated the significance of HMGB1 as DAMPs in the female reproductive system. These findings have shed light on the potential role of HMGB1 in the pathogenesis of diseases in female reproductive system and the possibilities of HMGB1-targeted therapies for treating them. Such therapies can help reduce inflammation and metabolic dysfunction and alleviate the symptoms of reproductive system diseases. Overall, the identification of HMGB1 as a key player in disease of the female reproductive system represents a significant breakthrough in our understanding of these conditions and presents exciting opportunities for the development of novel therapies.
Collapse
Affiliation(s)
- Yu Ren
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Damin Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Xingxing Han
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Qiqi Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Beili Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Huijuan Zou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
36
|
Ferraz MDAMM, Ferronato GDA. Opportunities involving microfluidics and 3D culture systems to the in vitro embryo production. Anim Reprod 2023; 20:e20230058. [PMID: 37638255 PMCID: PMC10449241 DOI: 10.1590/1984-3143-ar2023-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/29/2023] [Indexed: 08/29/2023] Open
Abstract
Traditional methods of gamete handling, fertilization, and embryo culture often face limitations in efficiency, consistency, and the ability to closely mimic in vivo conditions. This review explores the opportunities presented by microfluidic and 3D culture systems in overcoming these challenges and enhancing in vitro embryo production. We discuss the basic principles of microfluidics, emphasizing their inherent advantages such as precise control of fluid flow, reduced reagent consumption, and high-throughput capabilities. Furthermore, we delve into microfluidic devices designed for gamete manipulation, in vitro fertilization, and embryo culture, highlighting innovations such as droplet-based microfluidics and on-chip monitoring. Next, we explore the integration of 3D culture systems, including the use of biomimetic scaffolds and organ-on-a-chip platforms, with a particular focus on the oviduct-on-a-chip. Finally, we discuss the potential of these advanced systems to improve embryo production outcomes and advance our understanding of early embryo development. By leveraging the unique capabilities of microfluidics and 3D culture systems, we foresee significant advancements in the efficiency, effectiveness, and clinical success of in vitro embryo production.
Collapse
Affiliation(s)
- Marcia de Almeida Monteiro Melo Ferraz
- Faculty of Veterinary Medicine, Ludwig-Maximilians University of Munich, Oberschleißheim, Germany
- Gene Center, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Giuliana de Avila Ferronato
- Faculty of Veterinary Medicine, Ludwig-Maximilians University of Munich, Oberschleißheim, Germany
- Gene Center, Ludwig-Maximilians University of Munich, Munich, Germany
| |
Collapse
|
37
|
Massri N, Loia R, Sones JL, Arora R, Douglas NC. Vascular changes in the cycling and early pregnant uterus. JCI Insight 2023; 8:e163422. [PMID: 37288662 PMCID: PMC10393238 DOI: 10.1172/jci.insight.163422] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023] Open
Abstract
Uterine vascular remodeling is intrinsic to the cycling and early pregnant endometrium. Maternal regulatory factors such as ovarian hormones, VEGF, angiopoietins, Notch, and uterine natural killer cells significantly mediate these vascular changes. In the absence of pregnancy, changes in uterine vessel morphology and function correlate with different stages of the human menstrual cycle. During early pregnancy, vascular remodeling in rodents and humans results in decreased uterine vascular resistance and increased vascular permeability necessary for pregnancy success. Aberrations in these adaptive vascular processes contribute to increased risk of infertility, abnormal fetal growth, and/or preeclampsia. This Review comprehensively summarizes uterine vascular remodeling in the human menstrual cycle, and in the peri- and post-implantation stages in rodent species (mice and rats).
Collapse
Affiliation(s)
- Noura Massri
- Cell and Molecular Biology Graduate Program and
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Rachel Loia
- School of Graduate Studies, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| | - Jennifer L. Sones
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Ripla Arora
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
| | - Nataki C. Douglas
- Department of Obstetrics, Gynecology and Reproductive Health and
- Center for Immunity and Inflammation, Rutgers Biomedical and Health Sciences, Newark, New Jersey, USA
| |
Collapse
|
38
|
Kanter J, Gordon SM, Mani S, Sokalska A, Park JY, Senapati S, Huh DD, Mainigi M. Hormonal stimulation reduces numbers and impairs function of human uterine natural killer cells during implantation. Hum Reprod 2023; 38:1047-1059. [PMID: 37075311 PMCID: PMC10501469 DOI: 10.1093/humrep/dead069] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/05/2023] [Indexed: 04/21/2023] Open
Abstract
STUDY QUESTION How does an altered maternal hormonal environment, such as that seen during superovulation with gonadotropins in ART, impact human uterine immune cell distribution and function during the window of implantation? SUMMARY ANSWER Hormonal stimulation with gonadotropins alters abundance of maternal immune cells including uterine natural killer (uNK) cells and reduces uNK cell ability to promote extravillous trophoblast (EVT) invasion. WHAT IS KNOWN ALREADY An altered maternal hormonal environment, seen following ART, can lead to increased risk for adverse perinatal outcomes associated with disordered placentation. Maternal immune cells play an essential role in invasion of EVTs, a process required for proper establishment of the placenta, and adverse perinatal outcomes have been associated with altered immune cell populations. How ART impacts maternal immune cells and whether this can in turn affect implantation and placentation in humans remain unknown. STUDY DESIGN, SIZE, DURATION A prospective cohort study was carried out between 2018 and 2021 on 51 subjects: 20 from natural cycles 8 days after LH surge; and 31 from stimulated IVF cycles 7 days after egg retrieval. PARTICIPANTS/MATERIALS, SETTING, METHODS Endometrial biopsies and peripheral blood samples were collected during the window of implantation in subjects with regular menstrual cycles or undergoing superovulation. Serum estradiol and progesterone levels were measured by chemiluminescent competitive immunoassay. Immune cell populations in blood and endometrium were analyzed using flow cytometry. uNK cells were purified using fluorescence-activated cell sorting and were subjected to RNA sequencing (RNA-seq). Functional changes in uNK cells due to hormonal stimulation were evaluated using the implantation-on-a-chip (IOC) device, a novel bioengineered platform using human primary cells that mimics early processes that occur during pregnancy in a physiologically relevant manner. Unpaired t-tests, one-way ANOVA, and pairwise multiple comparison tests were used to statistically evaluate differences. MAIN RESULTS AND THE ROLE OF CHANCE Baseline characteristics were comparable for both groups. As expected, serum estradiol levels on the day of biopsy were significantly higher in stimulated (superovulated) patients (P = 0.0005). In the setting of superovulation, we found an endometrium-specific reduction in the density of bulk CD56+ uNK cells (P < 0.05), as well as in the uNK3 subpopulation (P = 0.025) specifically (CD103+ NK cells). In stimulated samples, we also found that the proportion of endometrial B cells was increased (P < 0.0001). Our findings were specific to the endometrium and not seen in peripheral blood. On the IOC device, uNK cells from naturally cycling secretory endometrium promote EVT invasion (P = 0.03). However, uNK cells from hormonally stimulated endometrium were unable to significantly promote EVT invasion, as measured by area of invasion, depth of invasion, and number of invaded EVTs by area. Bulk RNA-seq of sorted uNK cells from stimulated and unstimulated endometrium revealed changes in signaling pathways associated with immune cell trafficking/movement and inflammation. LIMITATIONS, REASONS FOR CAUTION Patient numbers utilized for the study were low but were enough to identify significant overall population differences in select immune cell types. With additional power and deeper immune phenotyping, we may detect additional differences in immune cell composition of blood and endometrium in the setting of hormonal stimulation. Flow cytometry was performed on targeted immune cell populations that have shown involvement in early pregnancy. A more unbiased approach might identify changes in novel maternal immune cells not investigated in this study. We performed RNA-seq only on uNK cells, which demonstrated differences in gene expression. Ovarian stimulation may also impact gene expression and function of other subsets of immune cells, as well as other cell types within the endometrium. Finally, the IOC device, while a major improvement over existing in vitro methods to study early pregnancy, does not include all possible maternal cells present during early pregnancy, which could impact functional effects seen. Immune cells other than uNK cells may impact invasion of EVTs in vitro and in vivo, though these remain to be tested. WIDER IMPLICATIONS OF THE FINDINGS These findings demonstrate that hormonal stimulation affects the distribution of uNK cells during the implantation window and reduces the proinvasive effects of uNK cells during early pregnancy. Our results provide a potential mechanism by which fresh IVF cycles may increase risk of disorders of placentation, previously linked to adverse perinatal outcomes. STUDY FUNDING/COMPETING INTEREST(S) Research reported in this publication was supported by the University of Pennsylvania University Research Funding (to M.M.), the Eunice Kennedy Shriver National Institute of Child Health and Human Development (P50HD068157 to M.M., S.S., and S.M.), National Center for Advancing Translational Sciences of the National Institutes of Health (TL1TR001880 to J.K.), the Institute for Translational Medicine and Therapeutics of the Perelman School of Medicine at the University of Pennsylvania, the Children's Hospital of Philadelphia Research Institute (to S.M.G.), and the National Institute of Allergy and Infectious Diseases (K08AI151265 to S.M.G.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. All authors declare no conflict of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- J Kanter
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S M Gordon
- Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - S Mani
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - A Sokalska
- Division of Reproductive Endocrinology and Infertility, Stanford University, Stanford, CA, USA
| | - J Y Park
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - S Senapati
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - D D Huh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - M Mainigi
- Division of Reproductive Endocrinology and Infertility, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
39
|
Krop J, Tian X, van der Hoorn ML, Eikmans M. The Mac Is Back: The Role of Macrophages in Human Healthy and Complicated Pregnancies. Int J Mol Sci 2023; 24:5300. [PMID: 36982375 PMCID: PMC10049527 DOI: 10.3390/ijms24065300] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/12/2023] Open
Abstract
Pregnancy is a fascinating immunological paradox: the semi-allogeneic fetus generally grows without any complications. In the placenta, fetal trophoblast cells come into contact with maternal immune cells. Inaccurate or inadequate adaptations of the maternal immune system could lead to problems with the functioning of the placenta. Macrophages are important for tissue homeostasis, cleanup, and the repair of damaged tissues. This is crucial for a rapidly developing organ such as the placenta. The consensus on macrophages at the maternal-fetal interface in pregnancy is that a major proportion have an anti-inflammatory, M2-like phenotype, that expresses scavenger receptors and is involved in tissue remodeling and the dampening of the immune reactions. Recent multidimensional analyses have contributed to a more detailed outlook on macrophages. The new view is that this lineage represents a highly diverse phenotype and is more prevalent than previously thought. Spatial-temporal in situ analyses during gestation have identified unique interactions of macrophages both with trophoblasts and with T cells at different trimesters of pregnancy. Here, we elaborate on the role of macrophages during early human pregnancy and at later gestation. Their possible effect is reviewed in the context of HLA incompatibility between mother and fetus, first in naturally conceived pregnancies, but foremost in pregnancies after oocyte donation. The potential functional consequences of macrophages for pregnancy-related immune reactions and the outcome in patients with recurrent pregnancy loss are also discussed.
Collapse
Affiliation(s)
- Juliette Krop
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Xuezi Tian
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marie-Louise van der Hoorn
- Department of Obstetrics and Gynaecology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michael Eikmans
- Department of Immunology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
40
|
Meakin C, Kim C, Lampert T, Aleksunes LM. High-throughput screening of toxicants that modulate extravillous trophoblast migration. Toxicol Lett 2023; 375:1-7. [PMID: 36535517 PMCID: PMC9877196 DOI: 10.1016/j.toxlet.2022.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Migration and subsequent invasion of extravillous trophoblasts into the uterus is essential for proper formation of the placenta. Disruption of these processes may result in poor pregnancy outcomes including preeclampsia, placenta accreta, fetal growth restriction, or fetal death. Currently, there are several methods for quantifying cell migration and invasion in vitro, each with limitations. Therefore, we developed a novel, high-throughput method to screen chemicals for their ability to alter human trophoblast migration. Human HTR8/SVneo trophoblast cells were cultured in Oris™ cell migration plates containing stopper barriers. After EVT cells attached and chemicals were added to media, stoppers were removed thereby creating a cell-free detection zone for migration. Entry of trophoblasts into this zone was monitored through imaging every 6 h and used to calculate a relative cell density. Chemicals known to increase (epidermal growth factor) and decrease (pertussis toxin and cadmium) trophoblast migration were used to validate this in vitro method. Next, a panel of environmental chemicals including bisphenols, mycoestrogens, and flame retardants, were screened for their ability to alter trophoblast invasion. In conclusion, a real-time method to track extravillous trophoblast migration offers potential for screening contaminants as placental toxicants.
Collapse
Affiliation(s)
- Cassandra Meakin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | - Christine Kim
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | | | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA; Environmental and Occupational Health Sciences Institute, Rutgers University, Piscataway, NJ, USA; Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
41
|
Zelinka-Khobzey MM, Tarasenko KV, Nesterenko LA, Starchenko II. MORPHOMETRIC CHARACTERISTICS OF PLACENTA IN WOMEN WITH PREECLAMPSIA AND OBESITY COMPARED TO WOMEN WITH NORMAL BODY WEIGHT. WIADOMOSCI LEKARSKIE (WARSAW, POLAND : 1960) 2023; 76:2593-2600. [PMID: 38290022 DOI: 10.36740/wlek202312107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
OBJECTIVE The aim: to investigate the morphometric characteristics of placentas in women with comorbidity of preeclampsia and obesity compared to women with physiological body weight, and to assess the efficacy of the prophylactic therapy course developed to prevent the occurrence of preeclampsia in pregnant women with obesity. PATIENTS AND METHODS Materials and methods: 25 biopsy samples of placental tissue were taken from women between 37 and 40 weeks of gestation with a physiological body weight and with class II obesity. The women were divided into five groups of five women in each: the 1st group included women with physiological body weight without obstetric and somatic pathology; the 2nd group involved women with physiological body weight, whose pregnancy was complicated with preeclampsia; the 3rd group was made up of women with class II obesity whose pregnancy was complicated with preeclampsia; the 4th group consisted of women with class II obesity, who received the special prophylactic therapy course, and the 5th group included women with class II obesity, who did not receive the prophylactic therapy course. RESULTS Results: The analysis of morphometric parameters of placenta samples taken from women with preeclampsia and obesity demonstrates a number of com-pensatory and adaptive changes in placenta under hypoxic conditions, and the most important of them include a significant decrease in the number and the mean diameter of the terminal villi, the reduction of volume of villious tree, an increase in the diameter of the capillaries of terminal villi. The morphometric parameters and histological structure in placenta samples from women with obesity, who received the special the prophylactic therapy course, as well as in placenta samples of the control group were similar to the gestational normative values. CONCLUSION Conclusions: The morphometric investigation of placenta samples taken from women with comorbidity of preeclampsia and obesity has shown a significant decrease in the mean diameter of the terminal villi and an increase in the diameter of the capillaries of these terminal villi when compared with a group of women with preeclampsia and physiological body weight. The study has also demonstrated the distortion of the percentage ratio of the volume of the intervil¬lous space and the ratio of medium-calibre villi. The combination of these changes indicates a lack of adaptive capabilities in the placenta during preeclampsia under increasing hypoxic condition.
Collapse
|
42
|
Fatmous M, Rai A, Poh QH, Salamonsen LA, Greening DW. Endometrial small extracellular vesicles regulate human trophectodermal cell invasion by reprogramming the phosphoproteome landscape. Front Cell Dev Biol 2022; 10:1078096. [PMID: 36619864 PMCID: PMC9813391 DOI: 10.3389/fcell.2022.1078096] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/02/2022] [Indexed: 12/24/2022] Open
Abstract
A series of cyclical events within the uterus are crucial for pregnancy establishment. These include endometrial regeneration following menses, under the influence of estrogen (proliferative phase), then endometrial differentiation driven by estrogen/progesterone (secretory phase), to provide a microenvironment enabling attachment of embryo (as a hatched blastocyst) to the endometrial epithelium. This is followed by invasion of trophectodermal cells (the outer layer of the blastocyst) into the endometrium tissue to facilitate intrauterine development. Small extracellular vesicles (sEVs) released by endometrial epithelial cells during the secretory phase have been shown to facilitate trophoblast invasion; however, the molecular mechanisms that underline this process remain poorly understood. Here, we show that density gradient purified sEVs (1.06-1.11 g/ml, Alix+ and TSG101+, ∼180 nm) from human endometrial epithelial cells (hormonally primed with estrogen and progesterone vs. estrogen alone) are readily internalized by a human trophectodermal stem cell line and promote their invasion into Matrigel matrix. Mass spectrometry-based proteome analysis revealed that sEVs reprogrammed trophectoderm cell proteome and their cell surface proteome (surfaceome) to support this invasive phenotype through upregulation of pro-invasive regulators associated with focal adhesions (NRP1, PTPRK, ROCK2, TEK), embryo implantation (FBLN1, NIBAN2, BSG), and kinase receptors (EPHB4/B2, ERBB2, STRAP). Kinase substrate prediction highlighted a central role of MAPK3 as an upstream kinase regulating target cell proteome reprogramming. Phosphoproteome analysis pinpointed upregulation of MAPK3 T204/T202 phosphosites in hTSCs following sEV delivery, and that their pharmacological inhibition significantly abrogated invasion. This study provides novel molecular insights into endometrial sEVs orchestrating trophoblast invasion, highlighting the microenvironmental regulation of hTSCs during embryo implantation.
Collapse
Affiliation(s)
- Monique Fatmous
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University (LTU), Melbourne, VIC, Australia
| | - Alin Rai
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Central Clinical School, Monash University, Melbourne, VIC, Australia,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia
| | - Qi Hui Poh
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia,Department of Biochemistry and Chemistry, LTU, Melbourne, VIC, Australia
| | - Lois A. Salamonsen
- Centre for Reproductive Health, Hudson Institute of Medical Research, Clayton, VIC, Australia,Department of Molecular and Translational Medicine, Monash University, Clayton, VIC, Australia
| | - David W. Greening
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Central Clinical School, Monash University, Melbourne, VIC, Australia,Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC, Australia,Baker Department of Cardiovascular Research, Translation and Implementation, LTU, Melbourne, VIC, Australia,Department of Biochemistry and Chemistry, LTU, Melbourne, VIC, Australia,*Correspondence: David W. Greening,
| |
Collapse
|
43
|
Ko G, Jeon TJ, Kim SM. Trophoblast Migration with Different Oxygen Levels in a Gel-Patterned Microfluidic System. MICROMACHINES 2022; 13:2216. [PMID: 36557515 PMCID: PMC9783522 DOI: 10.3390/mi13122216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/10/2022] [Accepted: 12/11/2022] [Indexed: 06/17/2023]
Abstract
In the placenta, substances such as nutrients, oxygen, and by-products are exchanged between the mother and the fetus, and the proper formation of the placenta determines the success of pregnancy, including the growth of the fetus. Preeclampsia is an obstetric disease in which the incomplete formation of the placenta occurs, which is known to occur when there is an abnormality in the invasion of trophoblast cells. The invasion of trophoblast cells is controlled by oxygen concentration, and HIF-1α changes according to oxygen concentration, showing a difference in cell mobility. MMP-2 and MMP-9 are observed to be high in the endometrium involved in trophoblast invasion, and the expression is regulated according to the oxygen concentration. In this experiment, cell culture was conducted using a gel-patterned system with a hypoxic chamber. Before the chip experiment, the difference in the expression of MMP-2 and MMP-9 according to the oxygen concentration was confirmed using a hypoxia chamber. After that, trophoblast cells (HTR8/SVneo) and endothelial cells (HUVECs) were separated and cultured through a physical barrier through a hydrogel on a microfluidic chip. Cells were cultured in a hypoxic chamber under controlled oxygen levels. It was confirmed that the mobility of trophoblast cells in culture on the chip was upregulated in a hypoxic environment through oxygen control. This suggests that the formation of a hypoxic environment in the endometrium where the invasion of trophoblast cells occurs plays a role in increasing cell mobility.
Collapse
Affiliation(s)
- Gun Ko
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Department of Biological Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Sun Min Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Department of Mechanical Engineering, Inha University, 100 Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
44
|
Cable J, Lutolf MP, Fu J, Park SE, Apostolou A, Chen S, Song CJ, Spence JR, Liberali P, Lancaster M, Meier AB, Pek NMQ, Wells JM, Capeling MM, Uzquiano A, Musah S, Huch M, Gouti M, Hombrink P, Quadrato G, Urenda JP. Organoids as tools for fundamental discovery and translation-a Keystone Symposia report. Ann N Y Acad Sci 2022; 1518:196-208. [PMID: 36177906 PMCID: PMC11293861 DOI: 10.1111/nyas.14874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Complex three-dimensional in vitro organ-like models, or organoids, offer a unique biological tool with distinct advantages over two-dimensional cell culture systems, which can be too simplistic, and animal models, which can be too complex and may fail to recapitulate human physiology and pathology. Significant progress has been made in driving stem cells to differentiate into different organoid types, though several challenges remain. For example, many organoid models suffer from high heterogeneity, and it can be difficult to fully incorporate the complexity of in vivo tissue and organ development to faithfully reproduce human biology. Successfully addressing such limitations would increase the viability of organoids as models for drug development and preclinical testing. On April 3-6, 2022, experts in organoid development and biology convened at the Keystone Symposium "Organoids as Tools for Fundamental Discovery and Translation" to discuss recent advances and insights from this relatively new model system into human development and disease.
Collapse
Affiliation(s)
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences (SV) and School of Engineering (STI), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science (SB), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Roche Institute for Translational Bioengineering (ITB), Pharma Research and Early Development (pRED), F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sunghee Estelle Park
- Department of Bioengineering and NSF Science and Technology Center for Engineering Mechanobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Athanasia Apostolou
- Emulate Inc, Boston, Massachusetts, USA
- Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Shuibing Chen
- Department of Surgery, Weill Cornell Medical College, New York City, New York, USA
| | - Cheng Jack Song
- Keck Medicine of University of Southern California, Los Angeles, California, USA
| | - Jason R Spence
- Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Prisca Liberali
- Friedrich Miescher Institute for Biomedical Research (FMI) and University of Basel, Basel, Switzerland
| | | | - Anna B Meier
- First Department of Medicine, Cardiology, Klinikum rechts der Isar, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Nicole Min Qian Pek
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Pulmonary Biology, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - James M Wells
- Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati, Ohio, USA
- Division of Developmental Biology and Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Meghan M Capeling
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, Michigan, USA
| | - Ana Uzquiano
- Department of Stem Cell and Regenerative Biology, Harvard University
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Samira Musah
- Developmental and Stem Cell Biology Program and Division of Nephrology, Department of Medicine and Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
- Center for Biomolecular and Tissue Engineering, Durham, North Carolina, USA
- Department of Biomedical Engineering, Pratt School of Engineering, Durham, North Carolina, USA
- Duke Regeneration Center, Duke University, Durham, North Carolina, USA
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Mina Gouti
- Stem Cell Modelling of Development & Disease Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Pleun Hombrink
- University Medical Center Utrecht and HUB Organoids, Utrecht, Netherlands
| | - Giorgia Quadrato
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| | - Jean-Paul Urenda
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine and Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
45
|
Alleviation of Severe Skin Insults Following High-Dose Irradiation with Isolated Human Fetal Placental Stromal Cells. Int J Mol Sci 2022; 23:ijms232113321. [DOI: 10.3390/ijms232113321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Skin exposure to high-dose irradiation, as commonly practiced in radiotherapy, affects the different skin layers, causing dry and wet desquamation, hyperkeratosis fibrosis, hard to heal wounds and alopecia and damaged hair follicles. Fetal tissue mesenchymal stromal cells (f-hPSC) were isolated from excised human fetal placental tissue, based on their direct migration from the tissue samples to the tissue dish. The current study follows earlier reports on for the mitigation of acute radiation syndrome following whole body high-dose exposure with remotely injected f-hPSC. Both the head only and a back skin flap of mice were irradiated with 16 &18 Gy, respectively, by 6MeV clinical linear accelerator electron beam. In both locations, the irradiated skin areas developed early and late radiation induced skin damages, including cutaneous fibrosis, lesions, scaring and severe hair follicle loss and reduced hair pigmentation. Injection of 2 × 106 f-hPSC, 3 and 8 weeks following 16 Gy head irradiation, and 1 and 4 weeks following the 18 Gy back skin only irradiation, resulted in significantly faster healing of radiation induced damages, with reduction of wet desquamation as measured by surface moisture level and minor recovery of the skin viscoelasticity. Detailed histological morphometry showed a clear alleviation of radiation induced hyperkeratosis in f-hPSC treated mice, with significant regain of hair follicles density. Following 16 Gy head irradiation, the hair follicles density in the scalp skin was reduced significantly by almost a half relative to the controls. A nearly full recovery of hair density was found in the f-hPSC treated mice. In the 18 Gy irradiated back skin, the hair follicles density dropped in a late stage by ~70% relative to naïve controls. In irradiated f-hPSC treated mice, it was reduced by only ~30% and was significantly higher than the non-treated group. Our results suggest that local injections of xenogeneic f-hPSC could serve as a simple, safe and highly effective non-autologous pro-regenerative treatment for high-dose radiation induced skin insults. We expect that such treatment could also be applied for other irradiated organs.
Collapse
|
46
|
Moffett A, Shreeve N. Local immune recognition of trophoblast in early human pregnancy: controversies and questions. Nat Rev Immunol 2022; 23:222-235. [PMID: 36192648 PMCID: PMC9527719 DOI: 10.1038/s41577-022-00777-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2022] [Indexed: 02/02/2023]
Abstract
The role of the maternal immune system in reproductive success in humans remains controversial. Here we focus on the events that occur in the maternal decidua during the first few weeks of human pregnancy, because this is the site at which maternal leukocytes initially interact with and can recognize fetal trophoblast cells, potentially involving allorecognition by both T cells and natural killer (NK) cells. NK cells are the dominant leukocyte population in first-trimester decidua, and genetic studies point to a role of allorecognition by uterine NK cells in establishing a boundary between the mother and the fetus. By contrast, definitive evidence that allorecognition by decidual T cells occurs during the first trimester is lacking. Thus, our view is that during the crucial period when the placenta is established, damaging T cell-mediated adaptive immune responses towards placental trophoblast are minimized, whereas NK cell allorecognition contributes to successful implantation and healthy pregnancy.
Collapse
Affiliation(s)
- Ashley Moffett
- grid.5335.00000000121885934Department of Pathology, University of Cambridge, Cambridge, UK
| | - Norman Shreeve
- grid.5335.00000000121885934Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Wen L, Tang F. Organoid research on human early development and beyond. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:512-523. [PMID: 37724162 PMCID: PMC10471100 DOI: 10.1515/mr-2022-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 09/20/2023]
Abstract
The organoid field has been developing rapidly during the last decade. Organoids for human pre-, peri- and post-implantation development have opened an avenue to study these biological processes in vitro, which have been hampered by lack of accessible research models for long term. The technologies of four fields, single cell omics sequencing, genome editing and lineage tracing, microfluidics and tissue engineering, have fueled the rapid development of the organoid field. In this review, we will discuss the organoid research on human early development as well as future directions of the organoid field combining with other powerful technologies.
Collapse
Affiliation(s)
- Lu Wen
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, P. R. China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Peking University, Beijing, P. R. China
| |
Collapse
|
48
|
James JL, Lissaman A, Nursalim YNS, Chamley LW. Modelling human placental villous development: designing cultures that reflect anatomy. Cell Mol Life Sci 2022; 79:384. [PMID: 35753002 PMCID: PMC9234034 DOI: 10.1007/s00018-022-04407-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/12/2022] [Accepted: 05/30/2022] [Indexed: 11/03/2022]
Abstract
The use of in vitro tools to study trophoblast differentiation and function is essential to improve understanding of normal and abnormal placental development. The relative accessibility of human placentae enables the use of primary trophoblasts and placental explants in a range of in vitro systems. Recent advances in stem cell models, three-dimensional organoid cultures, and organ-on-a-chip systems have further shed light on the complex microenvironment and cell-cell crosstalk involved in placental development. However, understanding each model's strengths and limitations, and which in vivo aspects of human placentation in vitro data acquired does, or does not, accurately reflect, is key to interpret findings appropriately. To help researchers use and design anatomically accurate culture models, this review both outlines our current understanding of placental development, and critically considers the range of established and emerging culture models used to study this, with a focus on those derived from primary tissue.
Collapse
Affiliation(s)
- Joanna L James
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Abbey Lissaman
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Yohanes N S Nursalim
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Lawrence W Chamley
- Department of Obstetrics and Gynaecology, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
49
|
Zou P, Yao J, Cui YN, Zhao T, Che J, Yang M, Li Z, Gao C. Advances in Cellulose-Based Hydrogels for Biomedical Engineering: A Review Summary. Gels 2022; 8:364. [PMID: 35735708 PMCID: PMC9222388 DOI: 10.3390/gels8060364] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/03/2022] [Accepted: 06/04/2022] [Indexed: 12/12/2022] Open
Abstract
In recent years, hydrogel-based research in biomedical engineering has attracted more attention. Cellulose-based hydrogels have become a research hotspot in the field of functional materials because of their outstanding characteristics such as excellent flexibility, stimulus-response, biocompatibility, and degradability. In addition, cellulose-based hydrogel materials exhibit excellent mechanical properties and designable functions through different preparation methods and structure designs, demonstrating huge development potential. In this review, we have systematically summarized sources and types of cellulose and the formation mechanism of the hydrogel. We have reviewed and discussed the recent progress in the development of cellulose-based hydrogels and introduced their applications such as ionic conduction, thermal insulation, and drug delivery. Also, we analyzed and highlighted the trends and opportunities for the further development of cellulose-based hydrogels as emerging materials in the future.
Collapse
Affiliation(s)
- Pengfei Zou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| | - Jiaxin Yao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| | - Ya-Nan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| | - Te Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang 050018, China
| | - Junwei Che
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian 271016, China
| | - Meiyan Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China; (P.Z.); (J.Y.); (Y.-N.C.); (T.Z.); (J.C.); (M.Y.)
| |
Collapse
|
50
|
Gauster M, Moser G, Wernitznig S, Kupper N, Huppertz B. Early human trophoblast development: from morphology to function. Cell Mol Life Sci 2022; 79:345. [PMID: 35661923 PMCID: PMC9167809 DOI: 10.1007/s00018-022-04377-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/13/2022] [Accepted: 05/13/2022] [Indexed: 12/18/2022]
Abstract
Human pregnancy depends on the proper development of the embryo prior to implantation and the implantation of the embryo into the uterine wall. During the pre-implantation phase, formation of the morula is followed by internalization of blastomeres that differentiate into the pluripotent inner cell mass lineage, while the cells on the surface undergo polarization and differentiate into the trophectoderm of the blastocyst. The trophectoderm mediates apposition and adhesion of the blastocyst to the uterine epithelium. These processes lead to a stable contact between embryonic and maternal tissues, resulting in the formation of a new organ, the placenta. During implantation, the trophectoderm cells start to differentiate and form the basis for multiple specialized trophoblast subpopulations, all of which fulfilling specific key functions in placentation. They either differentiate into polar cells serving typical epithelial functions, or into apolar invasive cells that adapt the uterine wall to progressing pregnancy. The composition of these trophoblast subpopulations is crucial for human placenta development and alterations are suggested to result in placenta-associated pregnancy pathologies. This review article focuses on what is known about very early processes in human reproduction and emphasizes on morphological and functional aspects of early trophoblast differentiation and subpopulations.
Collapse
Affiliation(s)
- Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Nadja Kupper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
| |
Collapse
|