1
|
Truong N, Zahra A, Lintao RCV, Chauhan R, Bento GF, Vidal Jr. M, Kim S, Lam PY, Conrads T, Conrads K, Han A, Menon R, Richardson LS. Modeling reproductive and pregnancy-associated tissues using organ-on-chip platforms: challenges, limitations, and the high throughput data frontier. Front Bioeng Biotechnol 2025; 13:1568389. [PMID: 40236940 PMCID: PMC11996799 DOI: 10.3389/fbioe.2025.1568389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Over the past decade, organ-on-chip technology (microphysiological systems or tissue chips) has reshaped in-vitro physiological and pathological modeling and pharmaceutical drug assessment. FDA Modernization Act 2.0 allows for alternatives to animal testing or the use of appropriate non-animal models/new approach methods (NAMs), such as Organ-on-chips (OC) platforms or in silico simulation models, to generate pre-clinical in-vitro drug trial data for regulatory purposes primes the microfluidic field to have exponential growth in the coming years. The changes in the approaches of regulatory agencies could significantly impact the development of therapeutics for use during pregnancy. However, limitations of the devices and molecular and biochemical assay shortfalls hinder the progress of the OOC field. This review describes available reproductive and pregnancy-related OOC platforms, and the current methodologies utilized to generate endpoint datasets (e.g., microscopic imaging, immunocytochemistry, real-time polymerase chain reaction, cytokine multiplex analysis). Microfluidic platform limitations, such as fewer number of cells or low supernatant volumes and restrictions regarding fabrication materials, are described. Novel approaches (e.g., spatial transcriptomics, imaging cytometry by time of flight, exosomes analysis using Exoview) to overcome these challenges are described. OOC platforms are primed to provide biologically relevant and clinically translational data that can revolutionize in-vitro physiological modeling, drug discovery, and toxicologic risk assessment. However, engineering adaptations to increase the throughput of devices (i.e., device arrays) and biological advancements to improve data throughput are both needed for these platforms to reach their full potential.
Collapse
Affiliation(s)
- Nina Truong
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Abir Zahra
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Ryan C. V. Lintao
- Institute of Reproductive Health, National Institutes of Health, University of the Philippines Manila, Manila, Philippines
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Rahul Chauhan
- John Sealy School of Medicine, University Blvd., Galveston, TX, United States
| | - Giovana Fernanda Bento
- Department of Pathology, Botucatu Medical School, São Paulo State University, São Paulo, Brazil
| | - Manuel Vidal Jr.
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
- College of Medicine, San Beda University, Manila, Philippines
- Department of Chemistry, College of Science, De La Salle University Manila, Manila, Philippines
| | - Sungjin Kim
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Po Yi Lam
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Thomas Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Kelly Conrads
- Gynecologic Cancer Center of Excellence, Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
- Women’s Health Integrated Research Center, Women’s Service Line, Inova Health System, Falls Church, VA, United States
| | - Arum Han
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, United States
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - 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, United States
| |
Collapse
|
2
|
Li H, Zhang R. The role of calcium ions and the transient receptor potential vanilloid (TRPV) channel in bone remodelling and orthodontic tooth movement. Mol Biol Rep 2025; 52:297. [PMID: 40063148 DOI: 10.1007/s11033-025-10399-1] [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: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025]
Abstract
During orthodontic treatment, the application of orthodontic forces to the periodontal tissues leads to the activation of osteoblasts and osteoclasts, which in turn induces bone remodelling and tooth movement. Calcium is a biologically essential element that exists in the internal environment and cells as calcium ions(Ca2+). The concentration of extracellular Ca2+ can affect the activity and function of osteoblasts and osteoclasts, as well as regulate bone remodelling. In the cell, calcium ions play a crucial role in cell signal transduction, acting as a second messenger. The orthodontic force increases intracellular Ca2+ concentration through a series of cascade reactions that affect the differentiation and apoptosis of osteoblasts and osteoclasts. Calcium channels on the cell membrane are crucial for intracellular and extracellular calcium transport. Transient Receptor Potential Vanilloid (TRPV) is a calcium ion permeable and mechanosensitive receptor comprising six calcium channel subtypes, TRPV1-6. This review will focus on the crucial role of Ca2+ in bone metabolism and provide a comprehensive description of the function and mechanism of each specific TRPV channel subtype in orthodontic tooth movement and bone remodelling.
Collapse
Affiliation(s)
- Haoyu Li
- Orthodontic Department, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Fanjiacun Road 9, Fengtai District, Beijing, 100070, China
| | - Ruofang Zhang
- Orthodontic Department, School of Stomatology, Beijing Stomatological Hospital, Capital Medical University, Fanjiacun Road 9, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
3
|
Gehl AL, Klawitter D, Wissenbach U, Cole M, Wesely C, Löhr H, Weissgerber P, Sota A, Meyer MR, Fecher-Trost C. The proteomic landscape of trophoblasts unravels calcium-dependent syncytialization processes and beta-chorionic gonadotropin (ß-hCG) production. Reprod Biol Endocrinol 2025; 23:33. [PMID: 40038668 PMCID: PMC11877844 DOI: 10.1186/s12958-025-01362-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND The syncytiotrophoblast (STB) layer of the placenta is formed by cell fusion of cytotrophoblasts, acts as a feto-maternal barrier, is required for the production of pregnancy hormones such as chorionic gonadotropin, estradiol and progesterone and is also responsible for feto-maternal mineral exchange such as calcium. Adequate mineral supply and placental hormone production are essential for the maintenance of pregnancy, and disturbances in trophoblast integrity are associated with pregnancy complications. Since knowledge about the identity and expression levels of proteins in trophoblast and syncytiotrophoblast cells is limited so far, we analyzed the proteomes of trophoblast-like and syncytiotrophoblast-like BeWo cells under different calcium conditions. The investigation of protein expression profiles in combination with hormone assays can provide a better understanding of calcium-dependent cellular processes in trophoblasts and syncytiotrophoblasts. METHODS Here, we combine human trophoblast model cell cultures, hormone assays, antibody-based detection methods and high-resolution mass spectrometry analyzes to assess changes in cellular processes during syncytialization. RESULTS We monitored the changes in protein expression profiles during forskolin induced syncytialization of trophoblast-like cells in an unbiased manner and show that the expression of numerous proteins is strongly altered. Among them are enzymes of the glucocorticoid and sex hormones synthesis pathways such as cytochrome P450 (CYP) 19A1, CYP11A1, adrenodoxin (FDX1), hydroxysteroid dehydrogenase (HSD) 11β2 and HSD17β1, whose expression is strongly induced by syncytialization. The production of beta human chorionic gonadotropin (ß-hCG), progesterone and estradiol increase during syncytialization, while the secretion and synthesis of ß-hCG and the expression of several protein syncytiotrophoblast markers show a clear calcium dependence. CONCLUSION The broad applicability of semi-quantitative proteome profiling of cytotrophoblast- and syncytiotrophoblast-like cells provides new insights into signaling processes that occur in cytotrophoblasts /syncytiotrophoblasts during pregnancy.
Collapse
Affiliation(s)
- Anna-Lena Gehl
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Daniel Klawitter
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Ulrich Wissenbach
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Marnie Cole
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Christine Wesely
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Heidi Löhr
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Petra Weissgerber
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Adela Sota
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Markus R Meyer
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany
| | - Claudia Fecher-Trost
- Experimental and Clinical Pharmacology and Toxicology, Center for Molecular Signaling (PZMS), Saarland University, Buildings 61.4 and 46, 66421, Homburg, Germany.
| |
Collapse
|
4
|
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 2025; 25:979-995. [PMID: 39417748 DOI: 10.1039/d4lc00500g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
5
|
Wu X, Ye Z. Mechanoimmunology of T-Cell Activation. Scand J Immunol 2025; 101:e70009. [PMID: 39973081 DOI: 10.1111/sji.70009] [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: 08/28/2024] [Revised: 01/13/2025] [Accepted: 02/01/2025] [Indexed: 02/21/2025]
Abstract
T-cell activation, a pivotal process in the adaptive immune response, is initiated when the T cell receptor (TCR) recognises and binds to antigenic peptide-major histocompatibility complex (pMHC) molecules on the cell membrane. Emerging evidence indicates that mechanical cues regulate T-cell activation by modulating TCR signalling and mechanotransduction pathways, although the precise underlying mechanisms remain elusive. This review highlights recent findings suggesting that the TCR functions as a mechanosensor, capable of sensing and transmitting mechanical forces through conformational changes. Key steps in T-cell mechanotransduction are discussed, including the roles of the cytoskeleton, mechanosensitive channels such as Piezo 1 and microvilli in facilitating activation. Additionally, we analyse the mechanical responses of chimeric antigen receptor T cells. Understanding the mechanobiological mechanisms underlying T-cell activation offers novel insights and potential strategies for advancing immunotherapies and treating immune-related disorders.
Collapse
Affiliation(s)
- Xuelan Wu
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China
| | - Zhiyi Ye
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering, Chongqing, China
| |
Collapse
|
6
|
Walker V. The Molecular Biology of Placental Transport of Calcium to the Human Foetus. Int J Mol Sci 2025; 26:383. [PMID: 39796238 PMCID: PMC11720126 DOI: 10.3390/ijms26010383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
From fertilisation to delivery, calcium must be transported into and within the foetoplacental unit for intracellular signalling. This requires very rapid, precisely located Ca2+ transfers. In addition, from around the eighth week of gestation, increasing amounts of calcium must be routed directly from maternal blood to the foetus for bone mineralisation through a flow-through system, which does not impact the intracellular Ca2+ concentration. These different processes are mediated by numerous membrane-sited Ca2+ channels, transporters, and exchangers. Understanding the mechanisms is essential to direct interventions to optimise foetal development and postnatal bone health and to protect the mother and foetus from pre-eclampsia. Ethical issues limit the availability of human foetal tissue for study. Our insight into the processes of placental Ca2+ handling is advancing rapidly, enabled by developing genetic, analytical, and computer technology. Because of their diverse sources, the reports of new findings are scattered. This review aims to pull the data together and to highlight areas of uncertainty. Areas needing clarification include trafficking, membrane expression, and recycling of channels and transporters in the placental microvilli; placental metabolism of vitamin D in gestational diabetes and pre-eclampsia; and the vascular effects of increased endothelial Orai expression by pregnancy-specific beta-1-glycoproteins PSG1 and PSG9.
Collapse
Affiliation(s)
- Valerie Walker
- Department of Clinical Biochemistry, University Hospital Southampton NHS Foundation Trust, Southampton General Hospital, Southampton SO16 6YD, UK
| |
Collapse
|
7
|
Yoshie M, Tsuru A, Tamura K. [Roles of Trophoblast Differentiation, Cell Fusion, and Microvilli Formation in Placentation and Prospects for a Model for Their Assessment]. YAKUGAKU ZASSHI 2025; 145:49-52. [PMID: 39756925 DOI: 10.1248/yakushi.24-00174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
The placenta, which acts as an interface between fetal and maternal circulations, is an indispensable organ for fetal growth in mammalian pregnancy. It mediates the transportation of nutrients, the exchange of gases such as oxygen and carbon dioxide, and the excretion of waste products between the fetus and mother. The surface of placental villi is covered by two layers of mononuclear undifferentiated cytotrophoblasts (CT) and multinucleated syncytiotrophoblasts (ST). The formation of the multinucleated ST layer via fusion of CT is referred to as syncytialization, which is a well-characterized morphological sign of terminal differentiation. STs function not only as the placental barrier to separate maternal blood from fetal tissue but also as the main source of human chorionic gonadotropin (hCG) and progesterone (P4) during pregnancy. The significance of appropriate differentiation and fusion of CTs to form STs is demonstrated by the finding that disturbance of these processes is linked to the pathogenesis of pregnancy-associated complications such as hypertensive disorders of pregnancy (HDP) and fetal growth restriction (FGR). In this review, we focused on trophoblast differentiation, cell fusion and microvilli formation, and showed the role of short-chain fatty acid butyrate and progesterone receptor membrane component 1 (PGRMC1) in these processes. Furthermore, we described the evaluation of placental function and its prospects utilizing a quantitative trophoblast cell fusion system and microfluidic device.
Collapse
Affiliation(s)
- Mikihiro Yoshie
- Department of Endocrine Pharmacology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Atsuya Tsuru
- Department of Endocrine Pharmacology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| | - Kazuhiro Tamura
- Department of Endocrine Pharmacology, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
8
|
Zambuto SG, Scott AK, Oyen ML. Beyond 2D: Novel biomaterial approaches for modeling the placenta. Placenta 2024; 157:55-66. [PMID: 38514278 PMCID: PMC11399328 DOI: 10.1016/j.placenta.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/09/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
This review considers fully three-dimensional biomaterial environments of varying complexity as these pertain to research on the placenta. The developments in placental cell sources are first considered, along with the corresponding maternal cells with which the trophoblast interact. We consider biomaterial sources, including hybrid and composite biomaterials. Properties and characterization of biomaterials are discussed in the context of material design for specific placental applications. The development of increasingly complicated three-dimensional structures includes examples of advanced fabrication methods such as microfluidic device fabrication and 3D bioprinting, as utilized in a placenta context. The review finishes with a discussion of the potential for in vitro, three-dimensional placenta research to address health disparities and sexual dimorphism, especially in light of the exciting recent changes in the regulatory environment for in vitro devices.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Adrienne K Scott
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michelle L Oyen
- Department of Obstetrics and Gynecology, Washington University in St. Louis, St. Louis, MO, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Women's Health Engineering, Washington University in St. Louis, St. Louis, MO, USA; Center for Regenerative Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
9
|
Parameshwar PK, Vaillancourt C, Moraes C. Engineering placental trophoblast fusion: A potential role for biomechanics in syncytialization. Placenta 2024; 157:50-54. [PMID: 38448351 DOI: 10.1016/j.placenta.2024.02.006] [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: 11/06/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 03/08/2024]
Abstract
The process by which placental trophoblasts fuse to form the syncytiotrophoblast around the chorionic villi is not fully understood. Mechanical features of the in vivo and in vitro culture environments have recently emerged as having the potential to influence fusion efficiency, and considering these mechanical cues may ultimately allow predictive control of trophoblast syncytialization. Here, we review recent studies that suggest that biomechanical factors such as shear stress, tissue stiffness, and dimensionally-related stresses affect villous trophoblast fusion efficiency. We then discuss how these stimuli might arise in vivo and how they can be incorporated in cultures to study and enhance villous trophoblast fusion. We believe that this mechanical paradigm will provide novel insight into manipulating the syncytialization process to better engineer improved models, understand disease progression, and ultimately develop novel therapeutic strategies.
Collapse
Affiliation(s)
| | - Cathy Vaillancourt
- Institut National de la Recherche Scientifique (INRS)-Centre Armand-Frappier Santé Biotechnologie, Laval, QC, H7B 1B7, Canada; Department of Obstetrics and Gynecology, Université de Montréal, and Research Center Centre Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) du Nord-de-l'Île-de-Montréal, Montréal, QC, H3L 1K5, Canada
| | - Christopher Moraes
- Department of Biological and Biomedical Engineering, McGill University, Montréal, QC, H3A 2B4, Canada; Department of Chemical Engineering, McGill University, Montréal, QC, H3A 0C5, Canada; Goodman Cancer Research Centre, McGill University, Montréal, QC, H3A 1A3, Canada; Division of Experimental Medicine, McGill University, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
10
|
Ansel M, Ramachandran K, Dey G, Brunet T. Origin and evolution of microvilli. Biol Cell 2024; 116:e2400054. [PMID: 39233537 DOI: 10.1111/boc.202400054] [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: 05/17/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND INFORMATION Microvilli are finger-like, straight, and stable cellular protrusions that are filled with F-actin and present a stereotypical length. They are present in a broad range of cell types across the animal tree of life and mediate several fundamental functions, including nutrient absorption, photosensation, and mechanosensation. Therefore, understanding the origin and evolution of microvilli is key to reconstructing the evolution of animal cellular form and function. Here, we review the current state of knowledge on microvilli evolution and perform a bioinformatic survey of the conservation of genes encoding microvillar proteins in animals and their unicellular relatives. RESULTS We first present a detailed description of mammalian microvilli based on two well-studied examples, the brush border microvilli of enterocytes and the stereocilia of hair cells. We also survey the broader diversity of microvilli and discuss similarities and differences between microvilli and filopodia. Based on our bioinformatic survey coupled with carefully reconstructed molecular phylogenies, we reconstitute the order of evolutionary appearance of microvillar proteins. We document the stepwise evolutionary assembly of the "molecular microvillar toolkit" with notable bursts of innovation at two key nodes: the last common filozoan ancestor (correlated with the evolution of microvilli distinct from filopodia) and the last common choanozoan ancestor (correlated with the emergence of inter-microvillar adhesions). CONCLUSION AND SIGNIFICANCE We conclude with a scenario for the evolution of microvilli from filopodia-like ancestral structures in unicellular precursors of animals.
Collapse
Affiliation(s)
- Mylan Ansel
- Institut Pasteur, Université Paris-Cité, CNRS UMR3691, Evolutionary Cell Biology and Evolution of Morphogenesis Unit, Paris, France
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
- Master BioSciences, Département de Biologie, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Kaustubh Ramachandran
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Gautam Dey
- Cell Biology and Biophysics, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Thibaut Brunet
- Institut Pasteur, Université Paris-Cité, CNRS UMR3691, Evolutionary Cell Biology and Evolution of Morphogenesis Unit, Paris, France
| |
Collapse
|
11
|
Ebrahim S, Ballesteros A, Zheng WS, Mukherjee S, Hu G, Weng WH, Montgomery JS, Agyemang Y, Cui R, Sun W, Krystofiak E, Foster MP, Sotomayor M, Kachar B. Transmembrane channel-like 4 and 5 proteins at microvillar tips are potential ion channels and lipid scramblases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.22.609173. [PMID: 39229161 PMCID: PMC11370596 DOI: 10.1101/2024.08.22.609173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Microvilli-membrane bound actin protrusions on the surface of epithelial cells-are sites of critical processes including absorption, secretion, and adhesion. Increasing evidence suggests microvilli are mechanosensitive, but underlying molecules and mechanisms remain unknown. Here, we localize transmembrane channel-like proteins 4 and 5 (TMC4 and 5) and calcium and integrin binding protein 3 (CIB3) to microvillar tips in intestinal epithelial cells, near glycocalyx insertion sites. We find that TMC5 colocalizes with CIB3 in cultured cells and that a TMC5 fragment forms a complex with CIB3 in vitro. Homology and AlphaFold2 models reveal a putative ion permeation pathway in TMC4 and 5, and molecular dynamics simulations predict both proteins can conduct ions and perform lipid scrambling. These findings raise the possibility that TMC4 and 5 interact with CIB3 at microvillar tips to form a mechanosensitive complex, akin to TMC1 and 2, and CIB2 and 3, within the mechanotransduction channel complex at the tips of inner ear stereocilia.
Collapse
Affiliation(s)
- Seham Ebrahim
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Angela Ballesteros
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
- Section on Sensory Physiology and Biophysics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - W. Sharon Zheng
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Shounak Mukherjee
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Gaizun Hu
- Center for Membrane and Cell Physiology, Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA 22903, USA
| | - Wei-Hsiang Weng
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
| | - Jonathan S. Montgomery
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Yaw Agyemang
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Runjia Cui
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Willy Sun
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Evan Krystofiak
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mark P. Foster
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Marcos Sotomayor
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
- Biophysics Program, The Ohio State University, Columbus, OH 43210, USA
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, IL 60637, USA
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | - Bechara Kachar
- Laboratory of Cell Structure and Dynamics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Deng ZM, Dai FF, Wang RQ, Deng HB, Yin TL, Cheng YX, Chen GT. Organ-on-a-chip: future of female reproductive pathophysiological models. J Nanobiotechnology 2024; 22:455. [PMID: 39085921 PMCID: PMC11290169 DOI: 10.1186/s12951-024-02651-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 06/18/2024] [Indexed: 08/02/2024] Open
Abstract
The female reproductive system comprises the internal and external genitalia, which communicate through intricate endocrine pathways. Besides secreting hormones that maintain the female secondary sexual characteristics, it also produces follicles and offspring. However, the in vitro systems have been very limited in recapitulating the specific anatomy and pathophysiology of women. Organ-on-a-chip technology, based on microfluidics, can better simulate the cellular microenvironment in vivo, opening a new field for the basic and clinical research of female reproductive system diseases. This technology can not only reconstruct the organ structure but also emulate the organ function as much as possible. The precisely controlled fluidic microenvironment provided by microfluidics vividly mimics the complex endocrine hormone crosstalk among various organs of the female reproductive system, making it a powerful preclinical tool and the future of pathophysiological models of the female reproductive system. Here, we review the research on the application of organ-on-a-chip platforms in the female reproductive systems, focusing on the latest progress in developing models that reproduce the physiological functions or disease features of female reproductive organs and tissues, and highlighting the challenges and future directions in this field.
Collapse
Affiliation(s)
- Zhi-Min Deng
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Fang-Fang Dai
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Rui-Qi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Hong-Bing Deng
- Hubei International Scientific and Technological Cooperation Base of Sustainable Resource and Energy, Hubei Key Laboratory of Biomass Resource Chemistry and Environmental Biotechnology, School of Resource and Environmental Science, Wuhan University, Wuhan, Hubei, 430060, China
| | - Tai-Lang Yin
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Yan-Xiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| | - Gan-Tao Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China.
| |
Collapse
|
13
|
Kim S, Lam PY, Richardson LS, Menon R, Han A. A dynamic flow fetal membrane organ-on-a-chip system for modeling the effects of amniotic fluid motion. Biomed Microdevices 2024; 26:32. [PMID: 38963644 PMCID: PMC11624963 DOI: 10.1007/s10544-024-00714-1] [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] [Accepted: 06/27/2024] [Indexed: 07/05/2024]
Abstract
Fetal membrane (amniochorion), the innermost lining of the intrauterine cavity, surround the fetus and enclose amniotic fluid. Unlike unidirectional blood flow, amniotic fluid subtly rocks back and forth, and thus, the innermost amnion epithelial cells are continuously exposed to low levels of shear stress from fluid undulation. Here, we tested the impact of fluid motion on amnion epithelial cells (AECs) as a bearer of force impact and their potential vulnerability to cytopathologic changes that can destabilize fetal membrane functions. A previously developed amnion membrane (AM) organ-on-chip (OOC) was utilized but with dynamic flow to culture human fetal amnion membrane cells. The applied flow was modulated to perfuse culture media back and forth for 48 h to mimic fluid motion. A static culture condition was used as a negative control, and oxidative stress (OS) condition was used as a positive control representing pathophysiological changes. The impacts of fluidic motion were evaluated by measuring cell viability, cellular transition, and inflammation. Additionally, scanning electron microscopy (SEM) imaging was performed to observe microvilli formation. The results show that regardless of the applied flow rate, AECs and AMCs maintained their viability, morphology, innate meta-state, and low production of pro-inflammatory cytokines. E-cadherin expression and microvilli formation in the AECs were upregulated in a flow rate-dependent fashion; however, this did not impact cellular morphology or cellular transition or inflammation. OS treatment induced a mesenchymal morphology, significantly higher vimentin to cytokeratin 18 (CK-18) ratio, and pro-inflammatory cytokine production in AECs, whereas AMCs did not respond in any significant manner. Fluid motion and shear stress, if any, did not impact AEC cell function and did not cause inflammation. Thus, when using an amnion membrane OOC model, the inclusion of a dynamic flow environment is not necessary to mimic in utero physiologic cellular conditions of an amnion membrane.
Collapse
Affiliation(s)
- Sungjin Kim
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Po Yi Lam
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - 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
| | - 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
| | - Arum Han
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA.
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
14
|
Safarzadeh M, Richardson LS, Kammala AK, Mosebarger A, Bettayeb M, Kim S, Lam PY, Radnaa E, Han A, Menon R. A multi-organ, feto-maternal interface organ-on-chip, models pregnancy pathology and is a useful preclinical extracellular vesicle drug trial platform. EXTRACELLULAR VESICLE 2024; 3:100035. [PMID: 38872854 PMCID: PMC11175617 DOI: 10.1016/j.vesic.2024.100035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Pregnant women and their fetuses are often excluded from clinical trials due to missing drug-related pre-clinical trial information at the human feto-maternal interface (FMi). The two interfaces-placenta/decidua and fetal membranes/decidua are gatekeepers of drug transport; however, testing their functions is impractical during pregnancy. Limitations of current in-vivo/in-vitro models have hampered drug development and testing during pregnancy. Hence, major complications like preterm births and maternal and neonatal mortalities remain high. Advancements in organ-on-chip (OOC) platforms to test drug kinetics and efficacy and novel extracellular vesicle-based fetal drug delivery are expected to accelerate preclinical trials related to pregnancy complications. Here we report the development and testing of a humanized multi-organ fetal membrane/placenta (fetal)-decidua (maternal) interface OOC (FMi-PLA-OOC) that contains seven cell types interconnected through microchannels to maintain intercellular interactions as seen in-utero. Cytotoxicity, propagation, mechanism of action, and efficacy of engineered extracellular vesicles containing anti-inflammatory interleukin (IL)-10 (eIL-10) were evaluated to reduce FMi inflammation associated with preterm birth. A healthy and disease model (lipopolysaccharide-infectious inflammation) of the FMi-PLA-OOC was created and co-treated with eIL-10. eIL-10 propagated from the maternal to fetal side within 72-hours, localized in all cell types, showed no cytotoxicity, activated IL-10 signaling pathways, and reduced lipopolysaccharide-induced inflammation (minimized NF-kB activation and proinflammatory cytokine production). These data recapitulated eIL-10s' ability to reduce inflammation and delay infection-associated preterm birth in mouse models, suggesting FMi-PLA-OOC as an alternative approach to using animal models. Additionally, we report the utility of eIL-10 that can traverse through FMis to reduce inflammation-associated pregnancy complications.
Collapse
Affiliation(s)
- Melody Safarzadeh
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Lauren S. Richardson
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, 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, Texas, USA
| | - Angela Mosebarger
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Mohamed Bettayeb
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Sungjin Kim
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Po Yi Lam
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Enkhtuya Radnaa
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| | - Arum Han
- Department of Biomedical Engineering and Electrical and Computer Engineering, Texas A&M University, College Station, TX, USA
| | - Ramkumar Menon
- Division of Basic Science and Translational Research, Department of Obstetrics and Gynecology, The University of Texas Medical Branch at Galveston, Galveston, Texas, USA
| |
Collapse
|
15
|
Kim S, Lam PY, Richardson L, Menon R, Han A. A Dynamic Flow Fetal Membrane Organ-on-a-Chip System for Modeling the Effects of Amniotic Fluid Motion. RESEARCH SQUARE 2024:rs.3.rs-4372328. [PMID: 38798515 PMCID: PMC11118697 DOI: 10.21203/rs.3.rs-4372328/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Fetal membrane(amniochorion), the innermost lining of the intrauterine cavity, surround the fetus and enclose amniotic fluid. Unlike unidirectional blood flow, amniotic fluid subtly rocks back and forth, and thus, the innermost amnion epithelial cells are continuously exposed to low levels of shear stress from fluid undulation. Here, we tested the impact of fluid motion on amnion epithelial cells (AECs) as a bearer of force impact and their potential vulnerability to cytopathologic changes that can destabilize fetal membrane functions. An amnion membrane (AM) organ-on-chip (OOC) was utilized to culture human fetal amnion membrane cells. The applied flow was modulated to perfuse culture media back and forth for 48 hours flow culture to mimic fluid motion. Static culture condition was used as a negative control, and oxidative stress (OS) condition was used as a positive control for pathophysiological changes. The impacts of fluidic motion were evaluated by measuring cell viability, cellular transition, and inflammation. Additionally, scanning electron microscopy (SEM) imaging was performed to observe microvilli formation. The results show that regardless of the applied flow rate, AECs and AMCs maintained their viability, morphology, innate meta-state, and low production of pro-inflammatory cytokines. E-cadherin expression and microvilli formation in the AECs were upregulated in a flow rate-dependent fashion; however, this did not impact cellular morphology or cellular transition or inflammation. OS treatment induced a mesenchymal morphology, significantly higher vimentin to CK-18 ratio, and pro-inflammatory cytokine production in AECs, whereas AMCs did not respond in any significant manner. Fluid motion and shear stress, if any, did not impact AEC cell function and did not cause inflammation. Thus, when using an amnion membrane OOC model, the inclusion of a flow culture environment is not necessary to mimic any in utero physiologic cellular conditions of fetal membrane-derived cells.
Collapse
|
16
|
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
|
17
|
Lyssy F, Forstner D, Brugger BA, Ujčič K, Guettler J, Kupper N, Wernitznig S, Daxboeck C, Neuper L, El-Heliebi A, Kloimboeck T, Kargl J, Huppertz B, Ghaffari-Tabrizi-Wizsy N, Gauster M. The chicken chorioallantoic membrane assay revisited - A face-lifted approach for new perspectives in placenta research. Placenta 2024:S0143-4004(24)00113-9. [PMID: 38705802 DOI: 10.1016/j.placenta.2024.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/09/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
The study of very early human placentation is largely limited due to ethical restrictions on the use of embryonic tissue and the fact that the placental anatomy of common laboratory animal models varies considerably from that of humans. In recent years several promising models, including trophoblast stem cell-derived organoids, have been developed that have also proven useful for the study of important trophoblast differentiation processes. However, the consideration of maternal blood flow in trophoblast invasion models currently appears to be limited to animal models. An almost forgotten model to study the invasive behavior of trophoblasts is to culture them in vitro on the chicken chorioallantoic membrane (CAM), showing an extraembryonic vascular network in its mesenchymal stroma that is continuously perfused by the chicken embryonic blood circulation. Here, we present an extension of the previously described ex ovo CAM assay and describe the use of cavity-bearing trophoblast spheroids obtained from the first trimester cell line ACH-3P. We demonstrate how spheroids penetrated the CAM and that erosion of CAM vessels by trophoblasts led to filling of the spheroid cavities with chicken blood, mimicking initial steps of intervillous space blood perfusion. Moreover, we prove that this model is useful for state-of-the-art techniques including immunofluorescence and in situ padlock probe hybridization, making it a versatile tool to study aspects of trophoblast invasion in presence of blood flow.
Collapse
Affiliation(s)
- Freya Lyssy
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Beatrice A Brugger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Kaja Ujčič
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Nadja Kupper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Christine Daxboeck
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Teresa Kloimboeck
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Berthold Huppertz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | | | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria.
| |
Collapse
|
18
|
Wang Y, Xiong Y, Shi K, Effah CY, Song L, He L, Liu J. DNA nanostructures for exploring cell-cell communication. Chem Soc Rev 2024; 53:4020-4044. [PMID: 38444346 DOI: 10.1039/d3cs00944k] [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: 03/07/2024]
Abstract
The process of coordinating between the same or multiple types of cells to jointly execute various instructions in a controlled and carefully regulated environment is a very appealing field. In order to provide clearer insight into the role of cell-cell interactions and the cellular communication of this process in their local communities, several interdisciplinary approaches have been employed to enhance the core understanding of this phenomenon. DNA nanostructures have emerged in recent years as one of the most promising tools in exploring cell-cell communication and interactions due to their programmability and addressability. Herein, this review is dedicated to offering a new perspective on using DNA nanostructures to explore the progress of cell-cell communication. After briefly outlining the anchoring strategy of DNA nanostructures on cell membranes and the subsequent dynamic regulation of DNA nanostructures, this paper highlights the significant contribution of DNA nanostructures in monitoring cell-cell communication and regulating its interactions. Finally, we provide a quick overview of the current challenges and potential directions for the application of DNA nanostructures in cellular communication and interactions.
Collapse
Affiliation(s)
- Ya Wang
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Yamin Xiong
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Kangqi Shi
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Clement Yaw Effah
- The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Critical Care Medicine, Zhengzhou Key Laboratory of Sepsis, Henan Engineering Research Center for Critical Care Medicine, Zhengzhou 450003, China
| | - Lulu Song
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Leiliang He
- College of Public Health, Zhengzhou University, Zhengzhou 450001, China.
| | - Jianbo Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Key Laboratory for Bio-Nanotechnology and Molecular Engineering of Hunan Province, Hunan University, Changsha 410082, China.
| |
Collapse
|
19
|
Yang L, Liang P, Yang H, Coyne CB. Trophoblast organoids with physiological polarity model placental structure and function. J Cell Sci 2024; 137:jcs261528. [PMID: 37676312 PMCID: PMC10499031 DOI: 10.1242/jcs.261528] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/07/2023] [Indexed: 09/08/2023] Open
Abstract
Human trophoblast organoids (TOs) are a three-dimensional ex vivo culture model that can be used to study various aspects of placental development, physiology and pathology. However, standard culturing of TOs does not recapitulate the cellular orientation of chorionic villi in vivo given that the multi-nucleated syncytiotrophoblast (STB) develops largely within the inner facing surfaces of these organoids (STBin). Here, we developed a method to culture TOs under conditions that recapitulate the cellular orientation of chorionic villi in vivo. We show that culturing STBin TOs in suspension with gentle agitation leads to the development of TOs containing the STB on the outer surface (STBout). Using membrane capacitance measurements, we determined that the outermost surface of STBout organoids contain large syncytia comprising >50 nuclei, whereas STBin organoids contain small syncytia (<10 nuclei) and mononuclear cells. The growth of TOs under conditions that mimic the cellular orientation of chorionic villi in vivo thus allows for the study of a variety of aspects of placental biology under physiological conditions.
Collapse
Affiliation(s)
- Liheng Yang
- Department of Integrative Immunobiology, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Pengfei Liang
- Department of Biochemistry, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Huanghe Yang
- Department of Biochemistry, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Department of Neurobiology, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Carolyn B. Coyne
- Department of Integrative Immunobiology, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
20
|
Cenni V, Sabatelli P, Di Martino A, Merlini L, Antoniel M, Squarzoni S, Neri S, Santi S, Metti S, Bonaldo P, Faldini C. Collagen VI Deficiency Impairs Tendon Fibroblasts Mechanoresponse in Ullrich Congenital Muscular Dystrophy. Cells 2024; 13:378. [PMID: 38474342 PMCID: PMC10930931 DOI: 10.3390/cells13050378] [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: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/19/2024] [Indexed: 03/14/2024] Open
Abstract
The pericellular matrix (PCM) is a specialized extracellular matrix that surrounds cells. Interactions with the PCM enable the cells to sense and respond to mechanical signals, triggering a proper adaptive response. Collagen VI is a component of muscle and tendon PCM. Mutations in collagen VI genes cause a distinctive group of inherited skeletal muscle diseases, and Ullrich congenital muscular dystrophy (UCMD) is the most severe form. In addition to muscle weakness, UCMD patients show structural and functional changes of the tendon PCM. In this study, we investigated whether PCM alterations due to collagen VI mutations affect the response of tendon fibroblasts to mechanical stimulation. By taking advantage of human tendon cultures obtained from unaffected donors and from UCMD patients, we analyzed the morphological and functional properties of cellular mechanosensors. We found that the length of the primary cilia of UCMD cells was longer than that of controls. Unlike controls, in UCMD cells, both cilia prevalence and length were not recovered after mechanical stimulation. Accordingly, under the same experimental conditions, the activation of the Hedgehog signaling pathway, which is related to cilia activity, was impaired in UCMD cells. Finally, UCMD tendon cells exposed to mechanical stimuli showed altered focal adhesions, as well as impaired activation of Akt, ERK1/2, p38MAPK, and mechanoresponsive genes downstream of YAP. By exploring the response to mechanical stimulation, for the first time, our findings uncover novel unreported mechanistic aspects of the physiopathology of UCMD-derived tendon fibroblasts and point at a role for collagen VI in the modulation of mechanotransduction in tendons.
Collapse
Affiliation(s)
- Vittoria Cenni
- CNR-Institute of Molecular Genetics, via di Barbiano 1/10, 40136 Bologna, Italy (S.S.); (S.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Patrizia Sabatelli
- CNR-Institute of Molecular Genetics, via di Barbiano 1/10, 40136 Bologna, Italy (S.S.); (S.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Alberto Di Martino
- 1st Orthopedics and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (A.D.M.); (C.F.)
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy;
| | - Luciano Merlini
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy;
| | - Manuela Antoniel
- CNR-Institute of Molecular Genetics, via di Barbiano 1/10, 40136 Bologna, Italy (S.S.); (S.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Stefano Squarzoni
- CNR-Institute of Molecular Genetics, via di Barbiano 1/10, 40136 Bologna, Italy (S.S.); (S.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Simona Neri
- Medicine and Rheumatology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Spartaco Santi
- CNR-Institute of Molecular Genetics, via di Barbiano 1/10, 40136 Bologna, Italy (S.S.); (S.S.)
- IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
| | - Samuele Metti
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (S.M.); (P.B.)
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (S.M.); (P.B.)
| | - Cesare Faldini
- 1st Orthopedics and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (A.D.M.); (C.F.)
- Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy;
| |
Collapse
|
21
|
Inohaya A, Chigusa Y, Takakura M, Io S, Kim MA, Matsuzaka Y, Yasuda E, Ueda Y, Kawamura Y, Takamatsu S, Mogami H, Takashima Y, Mandai M, Kondoh E. Shear stress in the intervillous space promotes syncytial formation of iPS cells-derived trophoblasts†. Biol Reprod 2024; 110:300-309. [PMID: 37930227 DOI: 10.1093/biolre/ioad143] [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: 06/06/2023] [Revised: 10/01/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
The intervillous space of human placenta is filled with maternal blood, and villous trophoblasts are constantly exposed to the shear stress generated by maternal blood pressure and flow throughout the entire gestation period. However, the effects of shear stress on villous trophoblasts and their biological significance remain unknown. Here, using our recently established naïve human pluripotent stem cells-derived cytotrophoblast stem cells (nCTs) and a device that can apply arbitrary shear stress to cells, we investigated the impact of shear stress on early-stage trophoblasts. After 72 h of exposure to 10 dyn/cm2 shear stress, nCTs became fused and multinuclear, and mRNA expression of the syncytiotrophoblast (ST) markers, such as glial cell missing 1, endogenous retrovirus group W member 1 envelope, chorionic gonadotropin subunit beta 3, syndecan 1, pregnancy specific beta-1-glycoprotein 3, placental growth factor, and solute carrier family 2 member 1 were significantly upregulated compared to static conditions. Immunohistochemistry showed that shear stress increased fusion index, human chorionic gonadotropin secretion, and human placental lactogen secretion. Increased microvilli formation on the surface of nCTs under flow conditions was detected using scanning electron microscopy. Intracellular cyclic adenosine monophosphate significantly increased under flow conditions. Moreover, transcriptome analysis of nCTs subjected to shear stress revealed that shear stress upregulated ST-specific genes and downregulated CT-specific genes. Collectively, these findings indicate that shear stress promotes the differentiation of nCTs into ST.
Collapse
Affiliation(s)
- Asako Inohaya
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahito Takakura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shingo Io
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Min-A Kim
- Department of Obstetrics and Gynecology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Matsuzaka
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eriko Yasuda
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yusuke Ueda
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yosuke Kawamura
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shiro Takamatsu
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruta Mogami
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yasuhiro Takashima
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, Kyoto Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eiji Kondoh
- Department of Obstetrics and Gynecology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
22
|
Hori T, Okae H, Shibata S, Kobayashi N, Kobayashi EH, Oike A, Sekiya A, Arima T, Kaji H. Trophoblast stem cell-based organoid models of the human placental barrier. Nat Commun 2024; 15:962. [PMID: 38332125 PMCID: PMC10853531 DOI: 10.1038/s41467-024-45279-y] [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: 02/09/2022] [Accepted: 01/18/2024] [Indexed: 02/10/2024] Open
Abstract
Human placental villi have essential roles in producing hormones, mediating nutrient and waste exchange, and protecting the fetus from exposure to xenobiotics. Human trophoblast organoids that recapitulate the structure of villi could provide an important in vitro tool to understand placental development and the transplacental passage of xenobiotics. However, such organoids do not currently exist. Here we describe the generation of trophoblast organoids using human trophoblast stem (TS) cells. Following treatment with three kinds of culture medium, TS cells form spherical organoids with a single outer layer of syncytiotrophoblast (ST) cells that display a barrier function. Furthermore, we develop a column-type ST barrier model based on the culture condition of the trophoblast organoids. The bottom membrane of the column is almost entirely covered with syndecan 1-positive ST cells. The barrier integrity and maturation levels of the model are confirmed by measuring transepithelial/transendothelial electrical resistance (TEER) and the amount of human chorionic gonadotropin. Further analysis reveals that the model can be used to derive the apparent permeability coefficients of model compounds. In addition to providing a suite of tools for the study of placental development, our trophoblast models allow the evaluation of compound transfer and toxicity, which will facilitate drug development.
Collapse
Affiliation(s)
- Takeshi Hori
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Hiroaki Okae
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Shun Shibata
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Norio Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Eri H Kobayashi
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Akira Oike
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Asato Sekiya
- Department of Trophoblast Research, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 862-0973, Japan
| | - Takahiro Arima
- Department of Informative Genetics, Environment and Genome Research Center, Tohoku University Graduate School of Medicine, 2-1 Seiryo-cho, Aoba-ku, Sendai, 980-8575, Japan
| | - Hirokazu Kaji
- Department of Diagnostic and Therapeutic Systems Engineering, Institute of Biomaterials and Bioengineering (IBB), Tokyo Medical and Dental University (TMDU), 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo, 101-0062, Japan.
| |
Collapse
|
23
|
Sakai K, Miura S, Teshima TF, Goto T, Takeuchi S, Yamaguchi M. Small-artery-mimicking multi-layered 3D co-culture in a self-folding porous graphene-based film. NANOSCALE HORIZONS 2023; 8:1529-1536. [PMID: 37782508 DOI: 10.1039/d3nh00304c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
In vitro vessel-mimicking models have been spotlighted as a powerful tool for investigating cellular behaviours in vascular development and diseases. However, it is still challenging to create micro-scale tubular tissues while mimicking the structural features of small arteries. Here, we propose a 3D culture model of small vascular tissue using a self-folding graphene-based porous film. Vascular endothelial cells were encapsulated within the self-folding film to create a cellular construct with a controlled curvature radius ranging from 10 to 100 μm, which is comparable to the size of a human arteriole. Additionally, vascular endothelial cells and smooth muscle cells were separately co-cultured on the inner and outer surfaces of the folded film, respectively. The porous wall worked as a permeable barrier between them, affecting the cell-cell communications like the extracellular layer in the artery wall. Thus, the culture model recapitulates the structural features of a small artery and will help us better understand intercellular communications at the artery wall in physiological and pathological conditions.
Collapse
Affiliation(s)
- Koji Sakai
- NTT Basic Research Laboratories and Bio-Medical Informatics Research Center, NTT Corporation, 3-1 Morinosato Wakamiya, Atsugi, Kanagawa, 243-0198, Japan.
| | - Shigenori Miura
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Tetsuhiko F Teshima
- Medical and Health Informatics Laboratories, NTT Research Incorporated, 940 Stewart Dr, Sunnyvale, CA, 94085, USA
| | - Toichiro Goto
- NTT Basic Research Laboratories and Bio-Medical Informatics Research Center, NTT Corporation, 3-1 Morinosato Wakamiya, Atsugi, Kanagawa, 243-0198, Japan.
| | - Shoji Takeuchi
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
- Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masumi Yamaguchi
- NTT Basic Research Laboratories and Bio-Medical Informatics Research Center, NTT Corporation, 3-1 Morinosato Wakamiya, Atsugi, Kanagawa, 243-0198, Japan.
| |
Collapse
|
24
|
Li X, Li ZH, Wang YX, Liu TH. A comprehensive review of human trophoblast fusion models: recent developments and challenges. Cell Death Discov 2023; 9:372. [PMID: 37816723 PMCID: PMC10564767 DOI: 10.1038/s41420-023-01670-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/23/2023] [Accepted: 09/29/2023] [Indexed: 10/12/2023] Open
Abstract
As an essential component of the maternal-fetal interface, the placental syncytiotrophoblast layer contributes to a successful pregnancy by secreting hormones necessary for pregnancy, transporting nutrients, mediating gas exchange, balancing immune tolerance, and resisting pathogen infection. Notably, the deficiency in mononuclear trophoblast cells fusing into multinucleated syncytiotrophoblast has been linked to adverse pregnancy outcomes, such as preeclampsia, fetal growth restriction, preterm birth, and stillbirth. Despite the availability of many models for the study of trophoblast fusion, there exists a notable disparity from the ideal model, limiting the deeper exploration into the placental development. Here, we reviewed the existing models employed for the investigation of human trophoblast fusion from several aspects, including the development history, latest progress, advantages, disadvantages, scope of application, and challenges. The literature searched covers the monolayer cell lines, primary human trophoblast, placental explants, human trophoblast stem cells, human pluripotent stem cells, three-dimensional cell spheres, organoids, and placenta-on-a-chip from 1938 to 2023. These diverse models have significantly enhanced our comprehension of placental development regulation and the underlying mechanisms of placental-related disorders. Through this review, our objective is to provide readers with a thorough understanding of the existing trophoblast fusion models, making it easier to select most suitable models to address specific experimental requirements or scientific inquiries. Establishment and application of the existing human placental trophoblast fusion models.
Collapse
Affiliation(s)
- Xia Li
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
| | - Zhuo-Hang Li
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China
- Medical Laboratory Department, Traditional Chinese Medicine Hospital of Yaan, 625099, Sichuan, China
| | - Ying-Xiong Wang
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| | - Tai-Hang Liu
- Department of Bioinformatics, School of Basic Medical Sciences, Chongqing Medical University, 400016, Chongqing, China.
- The Joint International Research Laboratory of Reproduction and Development, Ministry of Education, 400016, Chongqing, China.
| |
Collapse
|
25
|
Patel K, Nguyen J, Shaha S, Brightwell A, Duan W, Zubkowski A, Domingo IK, Riddell M. Loss of polarity regulators initiates gasdermin-E-mediated pyroptosis in syncytiotrophoblasts. Life Sci Alliance 2023; 6:e202301946. [PMID: 37468163 PMCID: PMC10355286 DOI: 10.26508/lsa.202301946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
The syncytiotrophoblast is a human epithelial cell that is bathed in maternal blood on the maternal-facing surface of the human placenta. It therefore acts as a barrier and exchange interface between the mother and fetus. Syncytiotrophoblast dysfunction is a feature of pregnancy pathologies, like preeclampsia. Dysfunctional syncytiotrophoblasts display a loss of microvilli, which is a marker of aberrant apical-basal polarization, but little data exist about the regulation of syncytiotrophoblast polarity. Atypical PKC isoforms are conserved polarity regulators. Thus, we hypothesized that aPKC isoforms regulate syncytiotrophoblast polarity. Using human placental explant culture and primary trophoblasts, we found that loss of aPKC activity or expression induces syncytiotrophoblast gasdermin-E-dependent pyroptosis, a form of programmed necrosis. We also establish that TNF-α induces an isoform-specific decrease in aPKC expression and gasdermin-E-dependent pyroptosis. Therefore, aPKCs are homeostatic regulators of the syncytiotrophoblast function and a pathogenically relevant pro-inflammatory cytokine leads to the induction of programmed necrosis at the maternal-fetal interface. Hence, our results have important implications for the pathobiology of placental disorders like preeclampsia.
Collapse
Affiliation(s)
- Khushali Patel
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada
| | - Jasmine Nguyen
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Sumaiyah Shaha
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Amy Brightwell
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Wendy Duan
- Department of Physiology, University of Alberta, Edmonton, Canada
| | - Ashley Zubkowski
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
| | - Ivan K Domingo
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada
| | - Meghan Riddell
- Department of Physiology, University of Alberta, Edmonton, Canada
- Department of Obstetrics and Gynecology, University of Alberta, Edmonton, Canada
| |
Collapse
|
26
|
Przybylla R, Krohn M, Sellin ML, Frank M, Oswald S, Linnebacher M. Novel In Vitro Models for Cell Differentiation and Drug Transport Studies of the Human Intestine. Cells 2023; 12:2371. [PMID: 37830585 PMCID: PMC10572004 DOI: 10.3390/cells12192371] [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: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
The most common in vitro model for absorption, distribution, metabolism, and excretion (ADME) purposes is currently the Caco-2 cell line. However, clear differences in gene and protein expression towards the small intestine and an, at best, fair prediction accuracy of intestinal drug absorption restrict the usefulness of a model for intestinal epithelial cells. To overcome these limitations, we evaluated a panel of low-passaged patient-derived colorectal cancer cell lines of the HROC collection concerning similarities to small intestinal epithelial cells and their potential to predict intestinal drug absorption. After initial screening of a larger panel, ten cell lines with confluent outgrowth and long-lasting barrier-forming potential were further characterized in close detail. Tight junctional complexes and microvilli structures were detected in all lines, anda higher degree of differentiation was observed in 5/10 cell lines. All lines expressed multiple transporter molecules, with the expression levels in three lines being close to those of small intestinal epithelial cells. Compared with the Caco-2 model, three HROC lines demonstrated both higher similarity to jejunal epithelial tissue cells and higher regulatory potential of relevant drug transporters. In summary, these lines would be better-suited human small intestinal epithelium models for basic and translational research, especially for ADME studies.
Collapse
Affiliation(s)
- Randy Przybylla
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| | - Mathias Krohn
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| | - Marie-Luise Sellin
- Research Laboratory for Biomechanics and Implant Technology, Department of Orthopedics, Rostock University Medical Centre, 18057 Rostock, Germany;
| | - Marcus Frank
- Medical Biology and Electron Microscopy Centre, 18057 Rostock, Germany;
- Department Life, Light and Matter, University of Rostock, 18059 Rostock, Germany
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Centre, 18057 Rostock, Germany;
| | - Michael Linnebacher
- Molecular Oncology and Immunotherapy, Clinic of General Surgery, Rostock University Medical Centre, 18057 Rostock, Germany; (R.P.); (M.K.)
| |
Collapse
|
27
|
Brugger BA, Neuper L, Guettler J, Forstner D, Wernitznig S, Kummer D, Lyssy F, Feichtinger J, Krappinger J, El-Heliebi A, Bonstingl L, Moser G, Rodriguez-Blanco G, Bachkönig OA, Gottschalk B, Gruber M, Nonn O, Herse F, Verlohren S, Frank HG, Barapatre N, Kampfer C, Fluhr H, Desoye G, Gauster M. Fluid shear stress induces a shift from glycolytic to amino acid pathway in human trophoblasts. Cell Biosci 2023; 13:163. [PMID: 37684702 PMCID: PMC10492287 DOI: 10.1186/s13578-023-01114-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND The human placenta, a tissue with a lifespan limited to the period of pregnancy, is exposed to varying shear rates by maternal blood perfusion depending on the stage of development. In this study, we aimed to investigate the effects of fluidic shear stress on the human trophoblast transcriptome and metabolism. RESULTS Based on a trophoblast cell line cultured in a fluidic flow system, changes caused by shear stress were analyzed and compared to static conditions. RNA sequencing and bioinformatics analysis revealed an altered transcriptome and enriched gene ontology terms associated with amino acid and mitochondrial metabolism. A decreased GLUT1 expression and reduced glucose uptake, together with downregulated expression of key glycolytic rate-limiting enzymes, hexokinase 2 and phosphofructokinase 1 was observed. Altered mitochondrial ATP levels and mass spectrometry data, suggested a shift in energy production from glycolysis towards mitochondrial oxidative phosphorylation. This shift in energy production could be supported by increased expression of glutamic-oxaloacetic transaminase variants in response to shear stress as well as under low glucose availability or after silencing of GLUT1. The shift towards amino acid metabolic pathways could be supported by significantly altered amino acid levels, like glutamic acid, cysteine and serine. Downregulation of GLUT1 and glycolytic rate-limiting enzymes, with concomitant upregulation of glutamic-oxaloacetic transaminase 2 was confirmed in first trimester placental explants cultured under fluidic flow. In contrast, high fluid shear stress decreased glutamic-oxaloacetic transaminase 2 expression in term placental explants when compared to low flow rates. Placental tissue from pregnancies with intrauterine growth restriction are exposed to high shear rates and showed also decreased glutamic-oxaloacetic transaminase 2, while GLUT1 was unchanged and glycolytic rate-limiting enzymes showed a trend to be upregulated. The results were generated by using qPCR, immunoblots, quantification of immunofluorescent pictures, padlock probe hybridization, mass spectrometry and FRET-based measurement. CONCLUSION Our study suggests that onset of uteroplacental blood flow is accompanied by a shift from a predominant glycolytic- to an alternative amino acid converting metabolism in the villous trophoblast. Rheological changes with excessive fluidic shear stress at the placental surface, may disrupt this alternative amino acid pathway in the syncytiotrophoblast and could contribute to intrauterine growth restriction.
Collapse
Affiliation(s)
- Beatrice Anna Brugger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Lena Neuper
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Jacqueline Guettler
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Désirée Forstner
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Stefan Wernitznig
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Daniel Kummer
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Freya Lyssy
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Julian Krappinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Lilli Bonstingl
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Gerit Moser
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Giovanny Rodriguez-Blanco
- Clinical Institute for Medical and Chemical Laboratory Diagnosis, Medical University of Graz, Graz, Austria
| | - Olaf A Bachkönig
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Benjamin Gottschalk
- Division of Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria
| | - Michael Gruber
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
| | - Olivia Nonn
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefan Verlohren
- Department of Obstetrics and Gynaecology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Clinic for Obstetrics, Charité Berlin, Berlin, Germany
| | | | | | | | - Herbert Fluhr
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Martin Gauster
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, OST V, 8010, Graz, Austria.
| |
Collapse
|
28
|
Park JY, Lim H, Qin J, Lee LP. Creating mini-pregnancy models in vitro with clinical perspectives. EBioMedicine 2023; 95:104780. [PMID: 37657136 PMCID: PMC10480532 DOI: 10.1016/j.ebiom.2023.104780] [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: 05/29/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023] Open
Abstract
During the last decade, organs-on-chips or organoids microphysiological analysis platforms (MAP) have garnered attention in the practical applications of disease models, drug discovery, and developmental biology. Research on pregnant women has firm limitations due to ethical issues; thus, remodelling human pregnancy in vitro is highly beneficial for treatment modality development via disease remodelling or drug monitoring. This review highlights current efforts in bioengineering devices to reproduce human pregnancy and emphasises the significant convergence of biology, engineering, and maternal-foetal medicine. First, we review recent achievements in culturing cells from tissues involved in pregnancy; specifically, trophoblasts from the placenta. Second, we highlight developments in the reconstitution of pregnancy-related female reproductive organs across several structural and functional interpretations. Last, we examine research on the fundamental comprehension of pregnancy-associated diseases to find bioengineering solutions. Recreating human pregnancy through an engineered model is naturally complex; nevertheless, challenges are inevitable to progress precision medicine.
Collapse
Affiliation(s)
- Jee Yoon Park
- Department of Obstetrics and Gynecology, Seoul National University, Bundang Hospital, Seoul National University College of Medicine, Republic of Korea; Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA.
| | - Hosub Lim
- Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Luke P Lee
- Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Boston, MA, USA; Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, USA; Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea.
| |
Collapse
|
29
|
Lermant A, Rabussier G, Lanz HL, Davidson L, Porter IM, Murdoch CE. Development of a human iPSC-derived placental barrier-on-chip model. iScience 2023; 26:107240. [PMID: 37534160 PMCID: PMC10392097 DOI: 10.1016/j.isci.2023.107240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/28/2023] [Accepted: 06/26/2023] [Indexed: 08/04/2023] Open
Abstract
Although recently developed placenta-on-chip systems opened promising perspectives for placental barrier modeling, they still lack physiologically relevant trophoblasts and are poorly amenable to high-throughput studies. We aimed to implement human-induced pluripotent stem cells (hiPSC)-derived trophoblasts into a multi-well microfluidic device to develop a physiologically relevant and scalable placental barrier model. When cultured in a perfused micro-channel against a collagen-based matrix, hiPSC-derived trophoblasts self-arranged into a 3D structure showing invasive behavior, fusogenic and endocrine activities, structural integrity, and expressing placental transporters. RNA-seq analysis revealed that the microfluidic 3D environment boosted expression of genes related to early placental structural development, mainly involved in mechanosensing and cell surface receptor signaling. These results demonstrated the feasibility of generating a differentiated primitive syncytium from hiPSC in a microfluidic platform. Besides expanding hiPSC-derived trophoblast scope of applications, this study constitutes an important resource to improve placental barrier models and boost research and therapeutics evaluation in pregnancy.
Collapse
Affiliation(s)
- Agathe Lermant
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | | | | | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Iain M. Porter
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, DD1 5EH, UK
| | - Colin E. Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
30
|
Sung JH, Kim JJ. Recent advances in in vitro skin-on-a-chip models for drug testing. Expert Opin Drug Metab Toxicol 2023. [PMID: 37379024 DOI: 10.1080/17425255.2023.2227379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023]
Abstract
INTRODUCTION The skin is an organ that has the largest surface area and provides a barrier against external environment. While providing protection, it also interacts with other organs in the body and has implications in various diseases. Development of physiologically realistic in vitro models of the skin in the context of the whole body is important for studying these diseases, and will be a valuable tool for pharmaceutical, cosmetics, and food industry. AREA COVERED This article covers the basic background in skin structure, physiology, as well as drug metabolism in the skin, and dermatological diseases. We summarize various in vitro skin models currently available, and novel in vitro models based on organ-on-a-chip technology. We also explain the concept of multi-organ-on-a-chip and describe recent developments in this field aimed at recapitulating the interaction of the skin with other organs in the body. EXPERT OPINION Recent development in the organ-on-a-chip field has enabled the development of in vitro model systems that resemble human skin more closely than conventional models. In near future, we will be seeing various model systems that allow researchers to study complex diseases in a more mechanistic manner, which will help the development of new pharmaceuticals for such diseases.
Collapse
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| | - Jae Jung Kim
- Department of Chemical Engineering, Hongik University, Seoul, Republic of Korea
| |
Collapse
|
31
|
Wang X, Li N, Zhang Z, Qin K, Zhang H, Shao S, Liu B. Visualization of Cell Membrane Tension Regulated by the Microfilaments as a "Shock Absorber" in Micropatterned Cells. BIOLOGY 2023; 12:889. [PMID: 37372173 DOI: 10.3390/biology12060889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/17/2023] [Indexed: 06/29/2023]
Abstract
The extracellular stress signal transmits along the cell membrane-cytoskeleton-focal adhesions (FAs) complex, regulating the cell function through membrane tension. However, the mechanism of the complex regulating membrane tension is still unclear. This study designed polydimethylsiloxane stamps with specific shapes to change the actin filaments' arrangement and FAs' distribution artificially in live cells, visualized the membrane tension in real time, and introduced the concept of information entropy to describe the order degree of the actin filaments and plasma membrane tension. The results showed that the actin filaments' arrangement and FAs' distribution in the patterned cells were changed significantly. The hypertonic solution resulted in the plasma membrane tension of the pattern cell changing more evenly and slowly in the zone rich in cytoskeletal filaments than in the zone lacking filaments. In addition, the membrane tension changed less in the adhesive area than in the non-adhesive area when destroying the cytoskeletal microfilaments. This suggested that patterned cells accumulated more actin filaments in the zone where FAs were difficult to generate to maintain the stability of the overall membrane tension. The actin filaments act as shock absorbers to cushion the alternation in membrane tension without changing the final value of membrane tension.
Collapse
Affiliation(s)
- Xianmeng Wang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Na Li
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
- Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin 124221, China
| | - Kairong Qin
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
- Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Shuai Shao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
- Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| | - Bo Liu
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian 116024, China
- Liaoning Key Laboratory of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
32
|
Rabussier G, Bünter I, Bouwhuis J, Soragni C, van Zijp T, Ng CP, Domansky K, de Windt LJ, Vulto P, Murdoch CE, Bircsak KM, Lanz HL. Healthy and diseased placental barrier on-a-chip models suitable for standardized studies. Acta Biomater 2023; 164:363-376. [PMID: 37116636 DOI: 10.1016/j.actbio.2023.04.033] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 04/05/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
Pathologies associated with uteroplacental hypoxia, such as preeclampsia are among the leading causes of maternal and perinatal morbidity in the world. Its fundamental mechanisms are yet poorly understood due to a lack of good experimental models. Here we report an in vitro model of the placental barrier, based on co-culture of trophoblasts and endothelial cells against a collagen extracellular matrix in a microfluidic platform. The model yields a functional syncytium with barrier properties, polarization, secretion of relevant extracellular membrane components, thinning of the materno-fetal space, hormone secretion, and transporter function. The model is exposed to low oxygen conditions and perfusion flow is modulated to induce a pathological environment. This results in reduced barrier function, hormone secretion, and microvilli as well as an increased nuclei count, characteristics of preeclamptic placentas. The model is implemented in a titer plate-based microfluidic platform fully amenable to high-throughput screening. We thus believe this model could aid mechanistic understanding of preeclampsia and other placental pathologies associated with hypoxia/ischemia, as well as support future development of effective therapies through target and compound screening campaigns. STATEMENT OF SIGNIFICANCE: : The human placenta is a unique organ sustaining fetus growth but is also the source of severe pathologies, such as Preeclampsia. Though leading cause of perinatal mortality in the world, preeclampsia remains untreatable due to a lack of relevant in vitro placenta models. To better understand the pathology, we have developed 3D placental barrier models in a microfluidic device. The platform allows parallel culture of 40 perfused physiological miniaturized placental barriers, comprising a differentiated syncytium and endothelium that have been validated for transporter functions. Exposure to a hypoxic and ischemic environment enabled the mimicking of preeclamptic characteristics in high-throughput, which we believe could lead to a better understanding of the pathology as well as support future effective therapies development.
Collapse
Affiliation(s)
- Gwenaëlle Rabussier
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands; Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | - Ivan Bünter
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | | | - Camilla Soragni
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands; Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | | | - Chee Ping Ng
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | | | - Leon J de Windt
- Department of Cardiology, Maastricht University, Maastricht, 6226 ER, The Netherlands
| | - Paul Vulto
- MIMETAS BV, Oegstgeest, 2342 DH, The Netherlands
| | - Colin E Murdoch
- Systems Medicine, School of Medicine, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | | | | |
Collapse
|
33
|
Melatonin Supplementation during the Late Gestational Stage Enhances Reproductive Performance of Sows by Regulating Fluid Shear Stress and Improving Placental Antioxidant Capacity. Antioxidants (Basel) 2023; 12:antiox12030688. [PMID: 36978937 PMCID: PMC10045541 DOI: 10.3390/antiox12030688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
In this study, the effects of daily melatonin supplementation (2 mg/kg) at the late gestational stage on the reproductive performance of the sows have been investigated. This treatment potentially increased the litter size and birth survival rate and significantly increased the birth weight as well as the weaning weight and survival rate of piglets compared to the controls. The mechanistic studies have found that these beneficial effects of melatonin are not mediated by the alterations of reproductive hormones of estrogen and progesterone, nor did the glucose and lipid metabolisms, but they were the results of the reduced oxidative stress in placenta associated with melatonin supplementation. Indeed, the melatonergic system, including mRNAs and proteins of AANAT, MTNR1A and MTNR1B, has been identified in the placenta of the sows. The RNA sequencing of placental tissue and KEGG analysis showed that melatonin activated the placental tissue fluid shear stress pathway to stimulate the Nrf2 signaling pathway, which upregulated its several downstream antioxidant genes, including MGST1, GSTM3 and GSTA4, therefore, suppressing the placental oxidative stress. All these actions may be mediated by the melatonin receptor of MTNR1B.
Collapse
|
34
|
Tabrizi MEA, Gupta JK, Gross SR. Ezrin and Its Phosphorylated Thr567 Form Are Key Regulators of Human Extravillous Trophoblast Motility and Invasion. Cells 2023; 12:cells12050711. [PMID: 36899847 PMCID: PMC10000480 DOI: 10.3390/cells12050711] [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: 01/06/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/27/2023] Open
Abstract
The protein ezrin has been shown to enhance cancer cell motility and invasion leading to malignant behaviours in solid tumours, but a similar regulatory function in the early physiological reproduction state is, however, much less clear. We speculated that ezrin may play a key role in promoting first-trimester extravillous trophoblast (EVT) migration/invasion. Ezrin, as well as its Thr567 phosphorylation, were found in all trophoblasts studied, whether primary cells or lines. Interestingly, the proteins were seen in a distinct cellular localisation in long, extended protrusions in specific regions of cells. Loss-of-function experiments were carried out in EVT HTR8/SVneo and Swan71, as well as primary cells, using either ezrin siRNAs or the phosphorylation Thr567 inhibitor NSC668394, resulting in significant reductions in both cell motility and cellular invasion, albeit with differences between the cells used. Our analysis further demonstrated that an increase in focal adhesion was, in part, able to explain some of the molecular mechanisms involved. Data collected using human placental sections and protein lysates further showed that ezrin expression was significantly higher during the early stage of placentation and, importantly, clearly seen in the EVT anchoring columns, further supporting the potential role of ezrin in regulating migration and invasion in vivo.
Collapse
Affiliation(s)
| | - Janesh K. Gupta
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
- Fetal Medicine Centre, Birmingham Women’s NHS Foundation Trust, Birmingham B15 2TT, UK
| | - Stephane R. Gross
- School of Life and Health Sciences, Aston University, Birmingham B4 7ET, UK
- Correspondence: ; Tel.: +44-0121-204-3467
| |
Collapse
|
35
|
Dusza HM, van Boxel J, van Duursen MBM, Forsberg MM, Legler J, Vähäkangas KH. Experimental human placental models for studying uptake, transport and toxicity of micro- and nanoplastics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 860:160403. [PMID: 36417947 DOI: 10.1016/j.scitotenv.2022.160403] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 06/16/2023]
Abstract
Micro- and nanoplastics (MNPs) are ubiquitous in the environment and have recently been found in human lungs, blood and placenta. However, data on the possible effects of MNPs on human health is extremely scarce. The potential toxicity of MNPs during pregnancy, a period of increased susceptibility to environmental insults, is of particular concern. The placenta provides a unique interface between maternal and fetal circulation which is essential for in utero survival and healthy pregnancy. Placental toxicokinetics and toxicity of MNPs are still largely unexplored and the limited studies performed up to now focus mainly on polystyrene particles. Practical and ethical considerations limit research options in humans, and extrapolation from animal studies is challenging due to marked differences between species. Nevertheless, diverse in vitro and ex vivo human placental models exist e.g., plasma membrane vesicles, mono-culture and co-culture of placental cells, placenta-on-a-chip, villous tissue explants, and placental perfusion that can be used to advance this research area. The objective of this concise review is to recapitulate different human placental models, summarize the current understanding of placental uptake, transport and toxicity of MNPs and define knowledge gaps. Moreover, we provide perspectives for future research urgently needed to assess the potential hazards and risks of MNP exposure to maternal and fetal health.
Collapse
Affiliation(s)
- Hanna M Dusza
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Jeske van Boxel
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Majorie B M van Duursen
- Amsterdam Institute for Life and Environment, Faculty of Science, Vrije Universiteit Amsterdam, the Netherlands
| | - Markus M Forsberg
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Juliette Legler
- Division of Toxicology, Institute for Risk Assessment Sciences, Department of Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Kirsi H Vähäkangas
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
36
|
Multi-scale Modelling of Shear Stress on the Syncytiotrophoblast: Could Maternal Blood Flow Impact Placental Function Across Gestation? Ann Biomed Eng 2023; 51:1256-1269. [PMID: 36745293 PMCID: PMC10172261 DOI: 10.1007/s10439-022-03129-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 12/25/2022] [Indexed: 02/07/2023]
Abstract
The placenta is a critical fetal exchange organ, with a complex branching tree-like structure. Its surface is covered by a single multinucleated cell, the syncytiotrophoblast, which bathes in maternal blood for most of pregnancy. Mechanosensing protein expression by the syncytiotrophoblast at term suggests that shear stress exerted by maternal blood flow may modulate placental development and function. However, it is not known how the mechanosensitive capacity of the syncytiotrophoblast, or the shear stress it experiences, change across gestation. Here, we show that the syncytiotrophoblast expresses both mechanosensitive ion channels (Piezo 1, Polycystin 2, TRPV6) and motor proteins associated with primary cilia (Dynein 1, IFT88, Kinesin 2), with higher staining for all these proteins seen in late first trimester placentae than at term. MicroCT imaging of placental tissue was then used to inform computational models of blood flow at the placentone scale (using a porous media model), and at the villous scale (using explicit flow simulations). These two models are then linked to produce a combined model that allows the variation of shear stress across both these scales simultaneously. This combined model predicts that the range of shear stress on the syncytiotrophoblast is higher in the first-trimester than at term (0.8 dyne/cm2 median stress compared to 0.04 dyne/cm2) when considering both these scales. Together, this suggests that the nature of blood flow through the intervillous space, and the resulting shear stress on the syncytiotrophoblast have important influences on placental morphogenesis and function from early in pregnancy.
Collapse
|
37
|
Morales EA, Gaeta I, Tyska MJ. Building the brush border, one microvillus at a time. Curr Opin Cell Biol 2023; 80:102153. [PMID: 36827850 PMCID: PMC10033394 DOI: 10.1016/j.ceb.2023.102153] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/11/2023] [Accepted: 01/19/2023] [Indexed: 02/24/2023]
Abstract
Microvilli are actin bundle-supported surface protrusions assembled by diverse cell types to mediate biochemical and physical interactions with the external environment. Found on the surface of some of the earliest animal cells, primordial microvilli likely contributed to bacterial entrapment and feeding. Although millions of years of evolution have repurposed these protrusions to fulfill diverse roles such as detection of mechanical or visual stimuli in inner ear hair cells or retinal pigmented epithelial cells, respectively, solute uptake remains a key essential function linked to these structures. In this mini review, we offer a brief overview of the composition and structure of epithelial microvilli, highlight recent discoveries on the growth of these protrusions early in differentiation, and point to fundamental questions surrounding microvilli biogenesis that remain open for future studies.
Collapse
Affiliation(s)
- E Angelo Morales
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Isabella Gaeta
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
38
|
Furuta A, Shima T, Yoshida-Kawaguchi M, Yamada K, Yasuda I, Tsuda S, Yamaki-Ushijima A, Yoneda S, Higashisaka K, Cheng SB, Matsumoto K, Tsutsumi Y, Sharma S, Saito S, Nakashima A. Chloroquine is a safe autophagy inhibitor for sustaining the expression of antioxidant enzymes in trophoblasts. J Reprod Immunol 2023; 155:103766. [PMID: 36470134 DOI: 10.1016/j.jri.2022.103766] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/19/2022] [Accepted: 11/22/2022] [Indexed: 11/27/2022]
Abstract
Inhibition of autophagy contributes to the pathophysiology of preeclampsia. Although chloroquine (CHQ) is an autophagy inhibitor, it can reduce the occurrence of preeclampsia in women with systemic lupus erythematosus. To clarify this important clinical question, this study aimed to address the safety of CHQ in trophoblast cells from the viewpoint of homeostasis, in which the anti-oxidative stress (OS) response and autophagy are involved. We used Western blotting to evaluate the protein levels in the trophoblast cells. The expression levels of heme oxygenase-1 (HO-1), an anti-OS enzyme, mediate resistance to OS induced by hydrogen peroxide (H2O2) in trophoblast cell lines. Among the autophagy modulators, bafilomycin A1 (BAF), an autophagy inhibitor, but not autophagy activators, suppressed HO-1 expression in BeWo cells; CHQ did not suppress HO-1 expression in BeWo cells. To clarify the role of autophagy in HO-1 induction, we observed no difference in HO-1 induction by H2O2 between autophagy-normal and autophagy-deficient cells. As for the mechanism of HO-1 induction by OS, BAF suppressed HO-1 induction by downregulating the expression of neighbor of BRCA1 gene 1 (NBR1) in the selective p62-NBR1-nuclear factor erythroid 2-related factor 2 (Nrf2) autophagy pathway. CHQ did not inhibit HO-1 expression by sustaining NBR1 expression in human villous tissues compared to BAF treatment. In conclusion, CHQ is a safer medicine than BAF for sustaining NBR1, which resist against OS in trophoblasts by connecting selective autophagy and the anti-OS response.
Collapse
Affiliation(s)
- Atsushi Furuta
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Tomoko Shima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Mihoko Yoshida-Kawaguchi
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Kiyotaka Yamada
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Ippei Yasuda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Sayaka Tsuda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Akemi Yamaki-Ushijima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Satoshi Yoneda
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan
| | - Kazuma Higashisaka
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shi-Bin Cheng
- Departments of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yasuo Tsutsumi
- Laboratory of Toxicology and Safety Science, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka 565-0871, Japan; The Center for Advanced Medical Engineering and Informatics, Osaka University, 1-6, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Surendra Sharma
- Departments of Pediatrics, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI 02905, USA
| | - Shigeru Saito
- University of Toyama, 3190 Gofuku, Toyama 9308555, Japan
| | - Akitoshi Nakashima
- Department of Obstetrics and Gynecology, Toyama Autophagy Team in Gynecology and Obstetrics, University of Toyama, 2630 Sugitani, Toyama 9300194, Japan.
| |
Collapse
|
39
|
Abdelkarim M, Perez-Davalos L, Abdelkader Y, Abostait A, Labouta HI. Critical design parameters to develop biomimetic organ-on-a-chip models for the evaluation of the safety and efficacy of nanoparticles. Expert Opin Drug Deliv 2023; 20:13-30. [PMID: 36440475 DOI: 10.1080/17425247.2023.2152000] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Organ-on-a-chip (OOC) models are based on microfluidics and can recapitulate the healthy and diseased microstructure of organs1 and tissues and the dynamic microenvironment inside the human body. However, the use of OOC models to evaluate the safety and efficacy of nanoparticles (NPs) is still in the early stages. AREAS COVERED The different design parameters of the microfluidic chip and the mechanical forces generated by fluid flow play a pivotal role in simulating the human environment. This review discusses the role of different key parameters on the performance of OOC models. These include the flow pattern, flow rate, shear stress (magnitude, rate, and distribution), viscosity of the media, and the microchannel dimensions and shape. We also discuss how the shear stress and other mechanical forces affect the transport of NPs across biological barriers, cell uptake, and their biocompatibility. EXPERT OPINION We describe several good practices and design parameters to consider for future OOC research. We submit that following these recommendations will help realize the full potential of the OOC models in the preclinical evaluation of novel therapies, including NPs.
Collapse
Affiliation(s)
- Mahmoud Abdelkarim
- Biomedical Engineering, University of Manitoba, R3T 5V6, Winnipeg, Manitoba, Canada.,College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Luis Perez-Davalos
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Yasmin Abdelkader
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada.,Department of Cell Biology, Biotechnology Research Institute, National Research Centre, 12622, Cairo, Egypt
| | - Amr Abostait
- College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada
| | - Hagar I Labouta
- Biomedical Engineering, University of Manitoba, R3T 5V6, Winnipeg, Manitoba, Canada.,College of Pharmacy, University of Manitoba, R3E 0T5, Winnipeg, Manitoba, Canada.,Children's Hospital Research Institute of Manitoba, R3E 3P4, Winnipeg, Manitoba, Canada.,Faculty of Pharmacy, Alexandria University, 21521, Alexandria, Egypt
| |
Collapse
|
40
|
Wei Y, Wang K, Xia Q, Li B, Liu L. 3D diversiform dynamic process of microvilli in living cells. Biochem Biophys Res Commun 2022; 635:114-119. [DOI: 10.1016/j.bbrc.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/17/2022] [Accepted: 10/01/2022] [Indexed: 11/25/2022]
|
41
|
Mosavati B, Oleinikov A, Du E. 3D microfluidics-assisted modeling of glucose transport in placental malaria. Sci Rep 2022; 12:15278. [PMID: 36088464 PMCID: PMC9464215 DOI: 10.1038/s41598-022-19422-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/29/2022] [Indexed: 11/10/2022] Open
Abstract
The human placenta is a critical organ, mediating the exchange of nutrients, oxygen, and waste products between fetus and mother. Placental malaria (PM) resulted from Plasmodium falciparum infections causes up to 200 thousand newborn deaths annually, mainly due to low birth weight, as well as 10 thousand mother deaths. In this work, a placenta-on-a-chip model is developed to mimic the nutrient exchange between the fetus and mother under the influence of PM. In this model, trophoblasts cells (facing infected or uninfected blood simulating maternal blood and termed “trophoblast side”) and human umbilical vein endothelial cells (facing uninfected blood simulating fetal blood and termed “endothelial” side) are cultured on the opposite sides of an extracellular matrix gel in a compartmental microfluidic system, forming a physiological barrier between the co-flow tubular structure to mimic a simplified maternal–fetal interface in placental villi. The influences of infected erythrocytes (IEs) sequestration through cytoadhesion to chondroitin sulfate A (CSA) expressed on the surface of trophoblast cells, a critical feature of PM, on glucose transfer efficiency across the placental barrier was studied. To create glucose gradients across the barrier, uninfected erythrocyte or IE suspension with a higher glucose concentration was introduced into the “trophoblast side” and a culture medium with lower glucose concentration was introduced into the “endothelial side”. The glucose levels in the endothelial channel in response to CSA-adherent erythrocytes infected with CS2 line of parasites in trophoblast channel under flow conditions was monitored. Uninfected erythrocytes served as a negative control. The results demonstrated that CSA-binding IEs added resistance to the simulated placental barrier for glucose perfusion and decreased the glucose transfer across this barrier. The results of this study can be used for better understanding of PM pathology and development of models useful in studying potential treatment of PM.
Collapse
|
42
|
Abostait A, Tyrrell J, Abdelkarim M, Shojaei S, Tse WH, El-Sherbiny IM, Keijzer R, Labouta HI. Placental Nanoparticle Uptake-On-a-Chip: The Impact of Trophoblast Syncytialization and Shear Stress. Mol Pharm 2022; 19:3757-3769. [PMID: 36053057 DOI: 10.1021/acs.molpharmaceut.2c00216] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The placenta is a dynamic and complex organ that plays an essential role in the health and development of the fetus. Placental disorders can affect the health of both the mother and the fetus. There is currently an unmet clinical need to develop nanoparticle-based therapies to target and treat placental disorders. However, little is known about the interaction of nanoparticles (NPs) with the human placenta under biomimetic conditions. Specifically, the impact of shear stress exerted on the trophoblasts (placental epithelial cells) by the maternal blood flow, the gradual fusion of the trophoblasts along the gestation period (syncytialization), and the impact of microvilli formation on the cell uptake of NPs is not known. To this end, we designed dynamic placenta-on-a-chip models using BeWo cells to recapitulate the micro-physiological environment, and we induced different degrees of syncytialization via chemical induction with forskolin. We characterized the degree of syncytialization quantitatively by measuring beta human chorionic gonadotropin (β-hCG) secretion, as well as qualitatively by immunostaining the tight junction protein, ZO-1, and counter nuclear staining. We also characterized microvilli formation under static and dynamic conditions via F-actin staining. We used these models to measure the cell uptake of chondroitin sulfate a binding protein (CSA) conjugated and control liposomes using confocal microscopy, followed by image analysis. Interestingly, exposure of the cells to a dynamic flow of media intrinsically induced syncytialization and microvilli formation compared to static controls. Under dynamic conditions, BeWo cells produced more β-hCG in conditions that increased the cell exposure time to forskolin (p < 0.005). Our cell uptake results clearly show a combined effect of the exerted shear stress and forskolin treatment on the cell uptake of liposomes as uptake increased in forskolin exposed conditions (p < 0.05). Overall, the difference in the extent of cell uptake of liposomes among the different conditions clearly displays a need for the development of dynamic models of the placenta that consider the changes in the placental cell phenotype along the gestation period, including syncytialization, microvilli formation, and the expression of different transport and uptake receptors. Knowledge generated from this work will inform future research aiming at developing drug delivery systems targeting the placenta.
Collapse
Affiliation(s)
- Amr Abostait
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada
| | - Jack Tyrrell
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Mahmoud Abdelkarim
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Biomedical Engineering, University of Manitoba, Winnipeg R3T 5V6, Canada
| | - Shahla Shojaei
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada
| | - Wai Hei Tse
- Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Depts of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg R3A 1R9, Canada
| | - Ibrahim M El-Sherbiny
- Nanomedicine Research Labs, Center for Materials Science, Zewail City of Science and Technology, Giza 12578, Egypt
| | - Richard Keijzer
- Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Depts of Surgery, Division of Pediatric Surgery, Pediatrics & Child Health and Physiology & Pathophysiology, University of Manitoba, Winnipeg R3A 1R9, Canada
| | - Hagar I Labouta
- College of Pharmacy, University of Manitoba, Winnipeg R3E 0T5, Canada.,Children's Hospital Research Institute of Manitoba, Winnipeg R3E 3P4, Canada.,Biomedical Engineering, University of Manitoba, Winnipeg R3T 5V6, Canada.,Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| |
Collapse
|
43
|
Sun L, Yeo T, Middha E, Gao Y, Lim CT, Watanabe S, Liu B. In Situ Visualization of Dynamic Cellular Effects of Phospholipid Nanoparticles via High-Speed Scanning Ion Conductance Microscopy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203285. [PMID: 35946985 DOI: 10.1002/smll.202203285] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/02/2022] [Indexed: 06/15/2023]
Abstract
Phospholipid nanoparticles have been actively employed for numerous biomedical applications. A key factor in ensuring effective and safe applications of these nanomaterials is the regulation of their interactions with target cells, which is significantly dependent on an in-depth understanding of the nanoparticle-cell interactions. To date, most studies investigating these nano-bio interactions have been performed under static conditions and may lack crucial real-time information. It is, however, noteworthy that the nanoparticle-cell interactions are highly dynamic. Consequently, to gain a deeper insight into the cellular effects of phospholipid nanoparticles, real-time observation of cellular dynamics after nanoparticle introduction is necessary. Herein, a proof-of-concept in situ visualization of the dynamic cellular effects of sub-100 nm phospholipid nanoparticles using high-speed scanning ion conductance microscopy (HS-SICM) is reported. It is revealed that upon introduction into the cellular environment, within a short timescale of hundreds of seconds, phospholipid nanoparticles can selectively modulate the edge motility and surface roughness of healthy fibroblast and cancerous epithelial cells. Furthermore, the dynamic deformation profiles of these cells can be selectively altered in the presence of phospholipid nanoparticles. This work is anticipated to further shed light on the real-time nanoparticle-cell interactions for improved formulation of phospholipid nanoparticles for numerous bioapplications.
Collapse
Affiliation(s)
- Linhao Sun
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Trifanny Yeo
- Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Eshu Middha
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| | - Yuji Gao
- Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology, National University of Singapore, MD6, 14 Medical Drive, Singapore, 117599, Singapore
- Department of Biomedical Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore, 117575, Singapore
| | - Shinji Watanabe
- Nano Life Science Institute (WPI-NanoLSI), Kanazawa University, Kakuma-machi, Kanazawa, 920-1192, Japan
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore, 117585, Singapore
| |
Collapse
|
44
|
Chevalier NR. Physical organogenesis of the gut. Development 2022; 149:276365. [DOI: 10.1242/dev.200765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
ABSTRACT
The gut has been a central subject of organogenesis since Caspar Friedrich Wolff’s seminal 1769 work ‘De Formatione Intestinorum’. Today, we are moving from a purely genetic understanding of cell specification to a model in which genetics codes for layers of physical–mechanical and electrical properties that drive organogenesis such that organ function and morphogenesis are deeply intertwined. This Review provides an up-to-date survey of the extrinsic and intrinsic mechanical forces acting on the embryonic vertebrate gut during development and of their role in all aspects of intestinal morphogenesis: enteric nervous system formation, epithelium structuring, muscle orientation and differentiation, anisotropic growth and the development of myogenic and neurogenic motility. I outline numerous implications of this biomechanical perspective in the etiology and treatment of pathologies, such as short bowel syndrome, dysmotility, interstitial cells of Cajal-related disorders and Hirschsprung disease.
Collapse
Affiliation(s)
- Nicolas R. Chevalier
- Laboratoire Matière et Systèmes Complexes, Université Paris Cité, CNRS UMR 7057 , 10 rue Alice Domon et Léonie Duquet, 75013 Paris , France
| |
Collapse
|
45
|
Menon R, Richardson L. Organ-on-a-chip for perinatal biology experiments. PLACENTA AND REPRODUCTIVE MEDICINE 2022; 1:98. [PMID: 36530581 PMCID: PMC9757604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cell culture and organ explant systems have traditionally been used by scientists in the reproductive biology and perinatal medicine area to address various research questions. Although most are unrelated to human pregnancy, animal models were also extensively used to study various mechanisms associated with pregnancy and parturition. However, limitations of traditional approaches have shifted the attention to the use of organ on a chip (OOC) technology. OOC platform simulates an organ using cells, and OOCs are biomimetic microfluidic systems comprising multiple cell types from an organ that mimic the environment of a physiological organ. OOC maintains intercellular interactions and helps to recreate organ physiology as expected for utero in perinatal medicine research. This short review introduces some basic concepts of OOC, and its utility based on some published reports.
Collapse
Affiliation(s)
- Ramkumar Menon
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555 TX, USA
| | - Lauren Richardson
- Department of Obstetrics & Gynecology, Division of Basic Science and Translational Research, The University of Texas Medical Branch at Galveston, Galveston 77555 TX, USA
| |
Collapse
|
46
|
Deng P, Cui K, Shi Y, Zhu Y, Wang Y, Shao X, Qin J. Fluidic Flow Enhances the Differentiation of Placental Trophoblast-Like 3D Tissue from hiPSCs in a Perfused Macrofluidic Device. Front Bioeng Biotechnol 2022; 10:907104. [PMID: 35845423 PMCID: PMC9280037 DOI: 10.3389/fbioe.2022.907104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
The human placenta serves as a multifunctional organ to maintain the proper development of a fetus. However, our knowledge of the human placenta is limited due to the lack of appropriate experimental models. In this work, we created an in vitro placental trophoblast-like model via self-organization of human induced pluripotent stem cells (hiPSCs) in a perfused 3D culture macrofluidic device. This device allowed cell seeding, in situ trophoblast lineage differentiation, and formation of trophoblast-like tissues from hiPSCs in a biomimetic microenvironment. It incorporated extracellular matrix (ECM) and fluid flow in a single device. After trophoblast lineage differentiation, we were able to generate the 3D clusters with major cell types of the human placenta, including trophoblast progenitor cytotrophoblasts (CTBs), differentiated subtypes, syncytiotrophoblasts (STBs), and extravillous trophoblasts (EVTs) under long-term 3D culture (∼23 days). Moreover, the formed tissues exhibited enhanced expressions of CTB-, STB-, and EVT-related markers at the level of genes and proteins under a dynamic culture compared with static conditions. RNA-seq analysis revealed the higher expression of trophoblast-specific genes in 3D tissues, indicating the essential role of fluid flow to promote the trophoblast differentiation of hiPSCs. The established placental 3D model combined a bioengineering strategy with developmental principles, providing a promising platform for the study of placental biology in a biomimetic microenvironment in health and disease.
Collapse
Affiliation(s)
- Pengwei Deng
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Kangli Cui
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Shi
- Dalian Key Laboratory of Reproduction and Mother-child Genetics, Dalian Women and Children’s Medical Group, Dalian, China
| | - Yujuan Zhu
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
| | - Yaqing Wang
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Xiaoguang Shao
- Dalian Key Laboratory of Reproduction and Mother-child Genetics, Dalian Women and Children’s Medical Group, Dalian, China
| | - Jianhua Qin
- Division of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Division of Biotechnology, Dalian Institute of Chemical Physics, University of Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
- *Correspondence: Jianhua Qin,
| |
Collapse
|
47
|
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
|
48
|
Zhang Y, Liang P, Yang L, Shan KZ, Feng L, Chen Y, Liedtke W, Coyne CB, Yang H. Functional coupling between TRPV4 channel and TMEM16F modulates human trophoblast fusion. eLife 2022; 11:e78840. [PMID: 35670667 PMCID: PMC9236608 DOI: 10.7554/elife.78840] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/05/2022] [Indexed: 11/15/2022] Open
Abstract
TMEM16F, a Ca2+-activated phospholipid scramblase (CaPLSase), is critical for placental trophoblast syncytialization, HIV infection, and SARS-CoV2-mediated syncytialization, however, how TMEM16F is activated during cell fusion is unclear. Here, using trophoblasts as a model for cell fusion, we demonstrate that Ca2+ influx through the Ca2+ permeable transient receptor potential vanilloid channel TRPV4 is critical for TMEM16F activation and plays a role in subsequent human trophoblast fusion. GSK1016790A, a TRPV4 specific agonist, robustly activates TMEM16F in trophoblasts. We also show that TRPV4 and TMEM16F are functionally coupled within Ca2+ microdomains in a human trophoblast cell line using patch-clamp electrophysiology. Pharmacological inhibition or gene silencing of TRPV4 hinders TMEM16F activation and subsequent trophoblast syncytialization. Our study uncovers the functional expression of TRPV4 and one of the physiological activation mechanisms of TMEM16F in human trophoblasts, thus providing us with novel strategies to regulate CaPLSase activity as a critical checkpoint of physiologically and disease-relevant cell fusion events.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Pengfei Liang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liheng Yang
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
| | - Ke Zoe Shan
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University Medical CentreDurhamUnited States
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua HospitalShanghaiChina
| | - Yong Chen
- Department of Neurology, Duke University Medical CenterDurhamUnited States
| | - Wolfgang Liedtke
- Department of Neurology, Duke University Medical CenterDurhamUnited States
- Department of Anesthesiology, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
- College of Dentistry, Department of Molecular Pathobiology, NYUNew YorkUnited States
| | - Carolyn B Coyne
- Department of Molecular Genetics and Microbiology, Duke University Medical CenterDurhamUnited States
- Duke Human Vaccine Institute, Duke UniversityDurhamUnited States
| | - Huanghe Yang
- Department of Biochemistry, Duke University Medical CenterDurhamUnited States
- Department of Neurobiology, Duke University Medical CenterDurhamUnited States
| |
Collapse
|
49
|
An improved in vitro model simulating the feto-maternal interface to study developmental effects of potentially toxic compounds: The example of titanium dioxide nanoparticles. Toxicol Appl Pharmacol 2022; 446:116056. [DOI: 10.1016/j.taap.2022.116056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/19/2022]
|
50
|
Khattar V, Wang L, Peng JB. Calcium selective channel TRPV6: Structure, function, and implications in health and disease. Gene 2022; 817:146192. [PMID: 35031425 PMCID: PMC8950124 DOI: 10.1016/j.gene.2022.146192] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/20/2021] [Accepted: 01/07/2022] [Indexed: 12/14/2022]
Abstract
Calcium-selective channel TRPV6 (Transient Receptor Potential channel family, Vanilloid subfamily member 6) belongs to the TRP family of cation channels and plays critical roles in transcellular calcium (Ca2+) transport, reuptake of Ca2+ into cells, and maintaining a local low Ca2+ environment for certain biological processes. Recent crystal and cryo-electron microscopy-based structures of TRPV6 have revealed mechanistic insights on how the protein achieves Ca2+ selectivity, permeation, and inactivation by calmodulin. The TRPV6 protein is expressed in a range of epithelial tissues such as the intestine, kidney, placenta, epididymis, and exocrine glands such as the pancreas, prostate and salivary, sweat, and mammary glands. The TRPV6 gene is a direct transcriptional target of the active form of vitamin D and is efficiently regulated to meet the body's need for Ca2+ demand. In addition, TRPV6 is also regulated by the level of dietary Ca2+ and under physiological conditions such as pregnancy and lactation. Genetic models of loss of function in TRPV6 display hypercalciuria, decreased bone marrow density, deficient weight gain, reduced fertility, and in some cases alopecia. The models also reveal that the channel plays an indispensable role in maintaining maternal-fetal Ca2+ transport and low Ca2+ environment in the epididymal lumen that is critical for male fertility. Most recently, loss of function mutations in TRPV6 gene is linked to transient neonatal hyperparathyroidism and early onset chronic pancreatitis. TRPV6 is overexpressed in a wide range of human malignancies and its upregulation is strongly correlated to tumor aggressiveness, metastasis, and poor survival in selected cancers. This review summarizes the current state of knowledge on the expression, structure, biophysical properties, function, polymorphisms, and regulation of TRPV6. The aberrant expression, polymorphisms, and dysfunction of this protein linked to human diseases are also discussed.
Collapse
Affiliation(s)
- Vinayak Khattar
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Lingyun Wang
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ji-Bin Peng
- Division of Nephrology, Department of Medicine, Nephrology Research and Training Center, Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|