1
|
Zambuto SG, Kolluru SS, Harley BAC, Oyen ML. Gelatin methacryloyl biomaterials and strategies for trophoblast research. Placenta 2024:S0143-4004(24)00659-3. [PMID: 39341721 DOI: 10.1016/j.placenta.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024]
Abstract
Rising maternal mortality rates in the U.S. are a significant public health issue that must be addressed; however, much of the basic science information required to target pregnancy-related pathologies have not yet been defined. Placental and blastocyst implantation research are challenging to perform in humans because of the early time frame of these processes in pregnancy and limited access to first trimester tissues. As a result, there is a critical need to develop model systems capable of studying these processes in increasing mechanistic detail. With the recent passing of the FDA Modernization Act 2.0 and advances in tissue engineering methods, three-dimensional microphysiological model systems offer an exciting opportunity to model early stages of placentation. Here, we detail the synthesis, characterization, and application of gelatin methacryloyl (GelMA) hydrogel platforms for studying trophoblast behavior in three-dimensional hydrogel systems. Photopolymerization strategies to fabricate GelMA hydrogels render the hydrogels homogeneous in terms of structure and stable under physiological temperatures, allowing for rigorous fabrication of reproducible hydrogel variants. Unlike other natural polymers that have minimal opportunity to tune their properties, GelMA hydrogel properties can be tuned across many axes of variation, including polymer degree of functionalization, gelatin bloom strength, light exposure time and intensity, polymer weight percent, photoinitiator concentration, and physical geometry. In this work, we aim to inspire and instruct the field to utilize GelMA biomaterial strategies for future placental research. With enhanced microphysiological models of pregnancy, we can now generate the basic science information required to address problems in pregnancy.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63130, USA; Dept. of Biomedical Engineering, USA; Center for Women's Health Engineering, USA.
| | - Samyuktha S Kolluru
- Center for Women's Health Engineering, USA; The Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| | - Brendan A C Harley
- Dept. Chemical and Biomolecular Engineering, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Michelle L Oyen
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63130, USA; Dept. of Biomedical Engineering, USA; Center for Women's Health Engineering, USA; The Institute of Materials Science & Engineering, Washington University in St. Louis, St. Louis, MO, 63130, USA
| |
Collapse
|
2
|
Kaviani S, Talebi A, Labbaf S, Karimzadeh F. Conductive GelMA/alginate/polypyrrole/graphene hydrogel as a potential scaffold for cardiac tissue engineering; Physiochemical, mechanical, and biological evaluations. Int J Biol Macromol 2024; 259:129276. [PMID: 38211921 DOI: 10.1016/j.ijbiomac.2024.129276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Cardiac failure can be a life-threatening condition that, if left untreated, can have significant consequences. Functional hydrogel has emerged as a promising platform for cardiac tissue engineering. In the proposed study, gelatin methacrylate (GelMA) and alginate, as a primary matrix to maintain cell viability and proliferation, and polypyrrole and carboxyl-graphene, to improve mechanical and electrical properties, are thoroughly evaluated. Initially, a polymer blend of GelMA/Alginate (1:1) was prepared, and then the addition of 2-5 wt% of polypyrrole was evaluated. Next, the effect of incorporating graphene-carboxyl nanosheets (1, 2, and 3 wt%) within the optimized scaffold with 2 wt% polypyrrole was thoroughly studied. The electrical conductivity of the hydrogels was significantly enhanced from 0.0615 ± 0.007 S/cm in GelMA/alginate to 0.124 ± 0.04 S/cm with the addition of 5 wt% polypyrrole. Also, 3 wt% carboxyl graphene improved the electrical conductivity to 0.27 ± 0.09 S/cm. The compressive strength of carboxyl-graphene-containing hydrogel was in the range of 175 to 520 kPa, and tensile strength was 61 and 133 kPa. The simplicity and low-cost fabrication, tunable mechanical properties, optimal electrical conductivity, blood compatibility, and non-cytotoxicity of GelMA/alginate/polypyrrole/graphene biocomposite hydrogel is a promising construct for cardiac tissue engineering.
Collapse
Affiliation(s)
- Sajedeh Kaviani
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Alireza Talebi
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Sheyda Labbaf
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran.
| | - Fathallah Karimzadeh
- Biomaterials Research Group, Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
3
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [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: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
4
|
Capella-Monsonís H, Cramer M, Turner N, Reing J, Zhang L, Kronengold RT, Bartolacci J, Badylak SF. The composition and mechanical properties of porcine placental ECM from three different breeds. Biomed Phys Eng Express 2023; 9:065012. [PMID: 37725946 DOI: 10.1088/2057-1976/acfb05] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/19/2023] [Indexed: 09/21/2023]
Abstract
Biologic scaffolds are extensively used in various clinical applications such as musculotendinous reconstruction, hernia repair or wound healing. Biologic scaffolds used in these applications vary in species, breed and tissue of origin, and other variables that affect their properties. Decellularization and sterilization processes also determine the characteristics of these scaffolds. The goal of the present study is to compare the composition and mechanical properties of decellularized porcine placental scaffolds from three different porcine breeds: Landrace, York and Duroc. Placental extracellular matrix (ECM) scaffolds from the three porcine breeds preserved the amnion/chorion ECM structure and the basement membrane markers laminin and collagen type IV. ECM placental scaffolds showed similar contents of collagen, elastin and lipids, and minimal differences in glycosaminoglycans content. Mechanical properties from the three breeds ECM placental scaffolds were also similar and stable for 24 months. While this study serves as preliminary characterization of porcine ECM scaffolds, future studies will determine their compatibility and suitability for tissue engineering applications.
Collapse
Affiliation(s)
- Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Madeline Cramer
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Neill Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Janet Reing
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Li Zhang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | | | - Joseph Bartolacci
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
| |
Collapse
|
5
|
Borodina E, Gzgzyan AM, Dzhemlikhanova LK, Niauri DA, Tolibova GK, Tral TG, Kogan IY, Safarian GK, Ostrinski Y, Churilov LP, Amital H, Blank M, Shoenfeld Y. Anti-TPO-mediated specific features of the placenta immunohistochemical profile and possible mechanisms for fetal loss. Clin Exp Immunol 2023; 213:235-242. [PMID: 37243348 PMCID: PMC10361736 DOI: 10.1093/cei/uxad057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/19/2023] [Accepted: 05/26/2023] [Indexed: 05/28/2023] Open
Abstract
Passive transfer of antithyroid antibodies in mice leads to reproductive disorders. The purpose was to assess the placental tissue of experimental animals under the influence of the circulating thyroperoxidase antibodies. We performed an immunohistochemical examination of murine placentae after a passive transfer of thyroperoxidase antibodies. Placentae of mice that passively transferred IgG from healthy donors were used as control samples. For histological examination, 30 placental samples were selected from mice from the anti-TPO group and 40 placental samples were taken from mice from the IgG group. Immunostaining for VEGFR1, THBS 1, Laminin, CD31, CD34, FGF-β, CD56, CD14, TNF-α, kisspeptin, MCL 1, and Annexin V was performed. There is a significant decrease in the relative area of the expression of VEGFR1 (23.42 ± 0.85 vs. 33.44 ± 0.35, P < 0.01), thrombospondin 1 (31.29 ± 0.83 vs. 34.51 ± 0.75, P < 0.01), CD14 (25.80 ± 0.57 vs. 32.07 ± 0.36, P < .01), CD56 (30.08 ± 0.90 vs. 34.92 ± 0.15, P < 0.01), kisspeptin (25.94 ± 0.47 vs. 31.27 ± 0.57, P < 0.01), MCL 1 (29.24 ± 1.06 vs. 38.57 ± 0.79, P < 0.01) in the labyrinth zone of the placentae of mice from the anti-TPO group compared with control group. A significant increase in the relative expression of laminin and FGF-β was noted in the group of mice to which antibodies to thyroperoxidase were transferred, compared with the control group (36.73 ± 1.38 vs. 29.83 ± 0.94, P < 0.01 and 23.26 ± 0.61 vs. 16.38 ± 1.01, P < 0.01respectively). Our study exposed an imbalance of pro- and anti-angiogenic factors, decreased representation of placental macrophages and NK cells, abnormal trophoblast invasion processes, and insufficient expression of antiapoptotic factors in the placentae of mice in which anti-TPO antibodies were passively transferred.
Collapse
Affiliation(s)
- Elena Borodina
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
| | - Alexander M Gzgzyan
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Liailia Kh Dzhemlikhanova
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Dariko A Niauri
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Gulrukhsor Kh Tolibova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Tatiana G Tral
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Igor Y Kogan
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Galina Kh Safarian
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, Saint Petersburg, Russia
| | - Yuri Ostrinski
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Leonid P Churilov
- Medical Faculty, Saint Petersburg State University, Saint Petersburg, Russia
| | - Howard Amital
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Miri Blank
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Affiliated with the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
6
|
Lopez E, Kamboj S, Chen C, Wang Z, Kellouche S, Leroy-Dudal J, Carreiras F, Lambert A, Aimé C. In Vitro Models of Ovarian Cancer: Bridging the Gap between Pathophysiology and Mechanistic Models. Biomolecules 2023; 13:biom13010103. [PMID: 36671488 PMCID: PMC9855568 DOI: 10.3390/biom13010103] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/23/2022] [Accepted: 12/25/2022] [Indexed: 01/06/2023] Open
Abstract
Ovarian cancer (OC) is a disease of major concern with a survival rate of about 40% at five years. This is attributed to the lack of visible and reliable symptoms during the onset of the disease, which leads over 80% of patients to be diagnosed at advanced stages. This implies that metastatic activity has advanced to the peritoneal cavity. It is associated with both genetic and phenotypic heterogeneity, which considerably increase the risks of relapse and reduce the survival rate. To understand ovarian cancer pathophysiology and strengthen the ability for drug screening, further development of relevant in vitro models that recapitulate the complexity of OC microenvironment and dynamics of OC cell population is required. In this line, the recent advances of tridimensional (3D) cell culture and microfluidics have allowed the development of highly innovative models that could bridge the gap between pathophysiology and mechanistic models for clinical research. This review first describes the pathophysiology of OC before detailing the engineering strategies developed to recapitulate those main biological features.
Collapse
Affiliation(s)
- Elliot Lopez
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Sahil Kamboj
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Changchong Chen
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Zixu Wang
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Sabrina Kellouche
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Johanne Leroy-Dudal
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe, EA1391, Groupe Matrice Extracellulaire et Physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, CEDEX, 95031 Neuville sur Oise, France
| | - Carole Aimé
- PASTEUR, Département de Chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
- Correspondence:
| |
Collapse
|
7
|
Mice Placental ECM Components May Provide A Three-Dimensional Placental Microenvironment. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 10:bioengineering10010016. [PMID: 36671588 PMCID: PMC9855196 DOI: 10.3390/bioengineering10010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Bioethical limitations impair deeper studies in human placental physiology, then most studies use human term placentas or murine models. To overcome these challenges, new models have been proposed to mimetize the placental three-dimensional microenvironment. The placental extracellular matrix plays an essential role in several processes, being a part of the establishment of materno-fetal interaction. Regarding these aspects, this study aimed to investigate term mice placental ECM components, highlighting its collagenous and non-collagenous content, and proposing a potential three-dimensional model to mimetize the placental microenvironment. For that, 18.5-day-old mice placenta, both control and decellularized (n = 3 per group) were analyzed on Orbitrap Fusion Lumos spectrometer (ThermoScientific) and LFQ intensity generated on MaxQuant software. Proteomic analysis identified 2317 proteins. Using ECM and cell junction-related ontologies, 118 (5.1%) proteins were filtered. Control and decellularized conditions had no significant differential expression on 76 (64.4%) ECM and cell junction-related proteins. Enriched ontologies in the cellular component domain were related to cell junction, collagen and lipoprotein particles, biological process domain, cell adhesion, vasculature, proteolysis, ECM organization, and molecular function. Enriched pathways were clustered in cell adhesion and invasion, and labyrinthine vasculature regulation. These preserved ECM proteins are responsible for tissue stiffness and could support cell anchoring, modeling a three-dimensional structure that may allow placental microenvironment reconstruction.
Collapse
|
8
|
Gonçalves BM, Graceli JB, da Rocha PB, Tilli HP, Vieira EM, de Sibio MT, Peghinelli VV, Deprá IC, Mathias LS, Olímpio RMC, Belik VC, Nogueira CR. Placental model as an important tool to study maternal-fetal interface. Reprod Toxicol 2022; 112:7-13. [PMID: 35714933 DOI: 10.1016/j.reprotox.2022.06.005] [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: 02/09/2022] [Revised: 05/29/2022] [Accepted: 06/09/2022] [Indexed: 10/18/2022]
Abstract
The placenta is a temporary organ that plays critical roles at the maternal-fetal interface. Normal development and function of the placenta is dependent on hormonal signaling pathways that make the placenta a target of endocrine disrupting chemical (EDC) action. Studies showing association between prenatal exposure, hormone disruption, and reproductive damage indicate that EDCs are developmentally toxic and can impact future generations. In this context, new placental models (trophoblast-derived cell lines, organotypic or 3D cell models, and physiologically based kinetic models) have been developed in order to create new approach methodology (NAM) to assess and even prevent such disastrous toxic harm in future generations. With the widespread discouragement of conducting animal studies, it has become irrefutable to develop in vitro models that can serve as a substitute for in vivo models. The goal of this review is to discuss the newest in vitro models to understand the maternal-fetal interface and predict placental development, physiology, and dysfunction generated by failures in molecular hormone control mechanisms, which, consequently, may change epigenetic programming to increase susceptibility to metabolic and other disorders in the offspring. We summarize the latest placental models for developmental toxicology studies, focusing mainly on three-dimensional (3D) culture models.
Collapse
Affiliation(s)
- Bianca M Gonçalves
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| | - Jones B Graceli
- Department of Morphology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitória, ES, Brazil
| | - Paula B da Rocha
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Helena P Tilli
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Ester M Vieira
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Maria T de Sibio
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Vinícius V Peghinelli
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Igor C Deprá
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Lucas S Mathias
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Regiane M C Olímpio
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Virgínia C Belik
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Célia R Nogueira
- Department of Clinical Medicine, Medical School, São Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
9
|
Qian X, Zhang Y. EZH2 enhances proliferation and migration of trophoblast cell lines by blocking GADD45A-mediated p38/MAPK signaling pathway. Bioengineered 2022; 13:12583-12597. [PMID: 35609316 PMCID: PMC9275956 DOI: 10.1080/21655979.2022.2074620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Impaired activity of the trophoblasts is a major contributor to the progression of pregnancy pathologies including preeclampsia (PE). This research probed the function of enhancer of zeste homolog 2 (EZH2) in activity of trophoblast cells and its correlation with growth arrest and DNA damage inducible alpha (GADD45A). EZH2 was predicted to be downregulated in placental tissues in PE according to a gene chip analysis, and reduced expression of EZH2 was detected in the placental tissues of patients with PE. Overexpression of EZH2 augmented proliferation and invasiveness of two trophoblast cell lines HTR-8/SVneo and JEG3 cells. EZH2 catalyzed trimethylation of lysine 27 on histone 3 (H3K27me3) in GADD45A promoter to suppress its transcription. GADD45A silencing increased the activity of the trophoblast cell lines and inactivated the p38/mitogen-activated protein kinase (MAPK) signaling pathway. Rescue experiments confirmed that either inhibition of GADD45A or p38 restored the proliferation, migration, and invasiveness of the trophoblast cell lines suppressed by EZH2 silencing. In conclusion, this work suggests that EZH2 enhances activity of trophoblast cell lines by suppressing GADD45A-mediated p38/MAPK signaling pathway.
Collapse
Affiliation(s)
- Xuefang Qian
- Department of Gynaecology and Obstetrics, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, P.R. China
| | - Yuying Zhang
- Department of Gynaecology and Obstetrics, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, P.R. China
| |
Collapse
|
10
|
Zambuto SG, Rattila S, Dveksler G, Harley BAC. Effects of Pregnancy-Specific Glycoproteins on Trophoblast Motility in Three-Dimensional Gelatin Hydrogels. Cell Mol Bioeng 2022; 15:175-191. [PMID: 35401843 PMCID: PMC8938592 DOI: 10.1007/s12195-021-00715-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/23/2021] [Indexed: 01/29/2023] Open
Abstract
Introduction Trophoblast invasion is a complex biological process necessary for establishment of pregnancy; however, much remains unknown regarding what signaling factors coordinate the extent of invasion. Pregnancy-specific glycoproteins (PSGs) are some of the most abundant circulating trophoblastic proteins in maternal blood during human pregnancy, with maternal serum concentrations rising to as high as 200-400 μg/mL at term. Methods Here, we employ three-dimensional (3D) trophoblast motility assays consisting of trophoblast spheroids encapsulated in 3D gelatin hydrogels to quantify trophoblast outgrowth area, viability, and cytotoxicity in the presence of PSG1 and PSG9 as well as epidermal growth factor and Nodal. Results We show PSG9 reduces trophoblast motility whereas PSG1 increases motility. Further, we assess bulk nascent protein production by encapsulated spheroids to highlight the potential of this approach to assess trophoblast response (motility, remodeling) to soluble factors and extracellular matrix cues. Conclusions Such models provide an important platform to develop a deeper understanding of early pregnancy.
Collapse
Affiliation(s)
- Samantha G. Zambuto
- grid.35403.310000 0004 1936 9991Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| | - Shemona Rattila
- grid.265436.00000 0001 0421 5525Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814 USA
| | - Gabriela Dveksler
- grid.265436.00000 0001 0421 5525Department of Pathology, Uniformed Services University of Health Sciences, Bethesda, MD 20814 USA
| | - Brendan A. C. Harley
- grid.35403.310000 0004 1936 9991Department Chemical and Biomolecular Engineering, Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews Ave, Urbana, IL 61801 USA ,grid.35403.310000 0004 1936 9991Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801 USA
| |
Collapse
|
11
|
Cherubini M, Erickson S, Haase K. Modelling the Human Placental Interface In Vitro-A Review. MICROMACHINES 2021; 12:884. [PMID: 34442506 PMCID: PMC8398961 DOI: 10.3390/mi12080884] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 07/17/2021] [Accepted: 07/24/2021] [Indexed: 12/29/2022]
Abstract
Acting as the primary link between mother and fetus, the placenta is involved in regulating nutrient, oxygen, and waste exchange; thus, healthy placental development is crucial for a successful pregnancy. In line with the increasing demands of the fetus, the placenta evolves throughout pregnancy, making it a particularly difficult organ to study. Research into placental development and dysfunction poses a unique scientific challenge due to ethical constraints and the differences in morphology and function that exist between species. Recently, there have been increased efforts towards generating in vitro models of the human placenta. Advancements in the differentiation of human induced pluripotent stem cells (hiPSCs), microfluidics, and bioprinting have each contributed to the development of new models, which can be designed to closely match physiological in vivo conditions. By including relevant placental cell types and control over the microenvironment, these new in vitro models promise to reveal clues to the pathogenesis of placental dysfunction and facilitate drug testing across the maternal-fetal interface. In this minireview, we aim to highlight current in vitro placental models and their applications in the study of disease and discuss future avenues for these in vitro models.
Collapse
Affiliation(s)
| | | | - Kristina Haase
- European Molecular Biology Laboratory (EMBL), 08003 Barcelona, Spain; (M.C.); (S.E.)
| |
Collapse
|
12
|
Winter M, Jankovic-Karasoulos T, Roberts CT, Bianco-Miotto T, Thierry B. Bioengineered Microphysiological Placental Models: Towards Improving Understanding of Pregnancy Health and Disease. Trends Biotechnol 2021; 39:1221-1235. [PMID: 33965246 DOI: 10.1016/j.tibtech.2021.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022]
Abstract
Driven by a lack of appropriate human placenta models, recent years have seen the introduction of bioengineered in vitro models to better understand placental health and disease. Thus far, the focus has been on the maternal-foetal barrier. However, there are many other physiologically and pathologically significant aspects of the placenta that would benefit from state-of-the-art bioengineered models, in particular, integrating advanced culture systems with contemporary biological concepts such as organoids. This critical review defines and discusses the key parameters required for the development of physiologically relevant in vitro models of the placenta. Specifically, it highlights the importance of cell type, mechanical forces, and culture microenvironment towards the use of physiologically relevant models to improve the understanding of human placental function and dysfunction.
Collapse
Affiliation(s)
- Marnie Winter
- ARC Centre of Excellence in Convergent BioNano Science and Technology and Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, South Australia, 5095, Australia.
| | - Tanja Jankovic-Karasoulos
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Claire T Roberts
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food, and Wine, University of Adelaide, Adelaide, South Australia, 5005, Australia; Robinson Research Institute, University of Adelaide, Adelaide, South Australia, 5005, Australia; Waite Research Institute, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Benjamin Thierry
- ARC Centre of Excellence in Convergent BioNano Science and Technology and Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, South Australia, 5095, Australia
| |
Collapse
|
13
|
Agnieray H, Glasson J, Chen Q, Kaur M, Domigan L. Recent developments in sustainably sourced protein-based biomaterials. Biochem Soc Trans 2021; 49:953-964. [PMID: 33729443 PMCID: PMC8106505 DOI: 10.1042/bst20200896] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 12/12/2022]
Abstract
Research into the development of sustainable biomaterials is increasing in both interest and global importance due to the increasing demand for materials with decreased environmental impact. This research field utilises natural, renewable resources to develop innovative biomaterials. The development of sustainable biomaterials encompasses the entire material life cycle, from desirable traits, and environmental impact from production through to recycling or disposal. The main objective of this review is to provide a comprehensive definition of sustainable biomaterials and to give an overview of the use of natural proteins in biomaterial development. Proteins such as collagen, gelatin, keratin, and silk, are biocompatible, biodegradable, and may form materials with varying properties. Proteins, therefore, provide an intriguing source of biomaterials for numerous applications, including additive manufacturing, nanotechnology, and tissue engineering. We give an insight into current research and future directions in each of these areas, to expand knowledge on the capabilities of sustainably sourced proteins as advanced biomaterials.
Collapse
Affiliation(s)
- H. Agnieray
- School of Chemical Sciences, University of Auckland, Auckland, New Zealand
| | - J.L. Glasson
- Department of Chemical and Material Engineering, University of Auckland, Auckland, New Zealand
| | - Q. Chen
- Department of Chemical and Material Engineering, University of Auckland, Auckland, New Zealand
| | - M. Kaur
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - L.J. Domigan
- Department of Chemical and Material Engineering, University of Auckland, Auckland, New Zealand
| |
Collapse
|
14
|
Choi E, Kim D, Kang D, Yang GH, Jung B, Yeo M, Park MJ, An S, Lee K, Kim JS, Kim JC, Jeong W, Yoo HH, Jeon H. 3D-printed gelatin methacrylate (GelMA)/silanated silica scaffold assisted by two-stage cooling system for hard tissue regeneration. Regen Biomater 2021; 8:rbab001. [PMID: 33738115 PMCID: PMC7955716 DOI: 10.1093/rb/rbab001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/23/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022] Open
Abstract
Among many biomaterials, gelatin methacrylate (GelMA), a photocurable protein, has been widely used in 3D bioprinting process owing to its excellent cellular responses, biocompatibility and biodegradability. However, GelMA still shows a low processability due to the severe temperature dependence of viscosity. To overcome this obstacle, we propose a two-stage temperature control system to effectively control the viscosity of GelMA. To optimize the process conditions, we evaluated the temperature of the cooling system (jacket and stage). Using the established system, three GelMA scaffolds were fabricated in which different concentrations (0, 3 and 10 wt%) of silanated silica particles were embedded. To evaluate the performances of the prepared scaffolds suitable for hard tissue regeneration, we analyzed the physical (viscoelasticity, surface roughness, compressive modulus and wettability) and biological (human mesenchymal stem cells growth, western blotting and osteogenic differentiation) properties. Consequently, the composite scaffold with greater silica contents (10 wt%) showed enhanced physical and biological performances including mechanical strength, cell initial attachment, cell proliferation and osteogenic differentiation compared with those of the controls. Our results indicate that the GelMA/silanated silica composite scaffold can be potentially used for hard tissue regeneration.
Collapse
Affiliation(s)
- Eunjeong Choi
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Dongyun Kim
- Department of Mechanical Engineering, Korea Polytechnic University, Sangidaehak-ro, Siheung, Gyeonggi-do 15073, South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| | - Bongsu Jung
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - MyungGu Yeo
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Min-Jeong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80, Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - SangHyun An
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - KyoungHo Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Jun Sik Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Jong Chul Kim
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu 41061, South Korea
| | - Woonhyeok Jeong
- Department of Plastic and Reconstructive Surgery, Dongsan Medical Center, Keimyung University College of Medicine, 1035 Dalgubeol-daero, Dalseo-gu, Daegu 42601, South Korea
| | - Hye Hyun Yoo
- Institute of Pharmaceutical Science and Technology, College of Pharmacy, Hanyang University, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-Do 15588, South Korea
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc, 55 Hanyangdaehak-Ro, Ansan, Gyeonggi-do 15588, South Korea
| |
Collapse
|
15
|
Zidan G, Greene CA, Etxabide A, Rupenthal ID, Seyfoddin A. Gelatine-based drug-eluting bandage contact lenses: Effect of PEGDA concentration and manufacturing technique. Int J Pharm 2021; 599:120452. [PMID: 33676990 DOI: 10.1016/j.ijpharm.2021.120452] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 02/25/2021] [Accepted: 02/27/2021] [Indexed: 01/21/2023]
Abstract
Drug-eluting bandage contact lenses (BCLs) have been widely studied as an alternative to eye drops due to their ability to increase the drug residence time and bioavailability as well as improve patient compliance. While silicone hydrogel polymers are commonly used in drug-eluting BCLs due to their transparency, mechanical properties and high oxygen permeability, gelatine hydrogels are also clear, flexible and have high oxygen permeability and may therefore be suitable contact lens materials. Moreover, the rheological properties of gelatine hydrogels allow their use as inks in extrusion-based 3D printers, therefore opening the door to a wide range of applications. Drug-loaded gelatine methacryloyl (GelMA) BCLs with different concentrations of poly (ethylene glycol) diacrylate (PEGDA) were prepared using solvent casting and 3D printing. The prepared lenses were characterised for their swelling ratio, in vitro degradation, and drug release properties. The results showed that the incorporation of 10% PEGDA improved the lenses' resistance to handling and protected them during degradation testing, reduced the swelling ratio and prolonged the release of dexamethasone (DEX). Both techniques were deemed suitable to use in the manufacturing of drug-eluting BCLs noting that the optimal formulation may vary according to the preparation technique utilised.
Collapse
Affiliation(s)
- Ghada Zidan
- Drug Delivery Research Group, School of Science, Auckland University of Technology, New Zealand
| | - Carol A Greene
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Alaitz Etxabide
- ALITEC Research Group, Department of Agronomy, Biotechnology and Food, School of Agricultural Engineering, Public University of Navarre (upna/nup), 31006 Pamplona-Iruña, Spain; School of Chemical Sciences 302, University of Auckland, 23 Symonds Street, Private Bag 92019, Auckland, New Zealand
| | - Ilva D Rupenthal
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Ali Seyfoddin
- Drug Delivery Research Group, School of Science, Auckland University of Technology, New Zealand.
| |
Collapse
|
16
|
Wheeler ML, Oyen ML. Bioengineering Approaches for Placental Research. Ann Biomed Eng 2021; 49:1805-1818. [PMID: 33420547 DOI: 10.1007/s10439-020-02714-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022]
Abstract
Research into the human placenta's complex functioning is complicated by a lack of suitable physiological in vivo models. Two complementary approaches have emerged recently to address these gaps in understanding, computational in silico techniques, including multi-scale modeling of placental blood flow and oxygen transport, and cellular in vitro approaches, including organoids, tissue engineering, and organ-on-a-chip models. Following a brief introduction to the placenta's structure and function and its influence on the substantial clinical problem of preterm birth, these different bioengineering approaches are reviewed. The cellular techniques allow for investigation of early first-trimester implantation and placental development, including critical biological processes such as trophoblast invasion and trophoblast fusion, that are otherwise very difficult to study. Similarly, computational models of the placenta and the pregnant pelvis at later-term gestation allow for investigations relevant to complications that occur when the placenta has fully developed. To fully understand clinical conditions associated with the placenta, including those with roots in early processes but that only manifest clinically at full-term, a holistic approach to the study of this fascinating, temporary but critical organ is required.
Collapse
Affiliation(s)
- Mackenzie L Wheeler
- Department of Engineering, East Carolina University, Greenville, NC, 27834, USA
| | - Michelle L Oyen
- Department of Engineering, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
17
|
Zhang H, Wu S, Ye S, Ma H, Liu Z. Preliminary RNA-microarray analysis of long non-coding RNA expression in abnormally invasive placenta. Exp Ther Med 2021; 21:13. [PMID: 33235622 PMCID: PMC7678644 DOI: 10.3892/etm.2020.9445] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/26/2020] [Indexed: 12/13/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are reported to have important roles in placental development and function, but the role of lncRNAs in abnormally invasive placenta (AIP) remains elusive. In the present study, the differential expression profiles of lncRNAs were analyzed to identify novel targets for further study of AIP. A total of 10 lncRNAs were chosen for validation by reverse transcription-quantitative PCR. To further determine the functions of dysregulated lncRNAs and their corresponding mRNAs, functional enrichment analysis, coexpression analysis were performed. A total of 329 lncRNAs and 179 mRNAs were identified to be differently expressed between the invasive and control group. Gene ontology analysis revealed that the 10 most significantly enriched functions included upregulated mRNAs and the most significantly enriched term was related to the proteinaceous extracellular matrix (ECM). In the pathway analysis, the two most significantly enriched pathways were the TGF-β signaling pathway for upregulated mRNAs and the pentose phosphate pathway for downregulated mRNAs. Furthermore, for certain dysregulated lncRNAs, their associated mRNAs were also dysregulated. Of note, BMP and activin membrane-bound inhibitor and TGF-β-induced, as the target genes of the TGF-β pathway, were indicated to be closely related to the ECM and invasive placental cells. Their nearby lncRNAs G008916 and vault RNA2-1 were also significantly dysregulated. In conclusion, significant lncRNAs with the potential to serve as biomarkers for AIP were identified.
Collapse
Affiliation(s)
- Huishan Zhang
- Department of Fetal Medicine Research, Foshan Fetal Medicine Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Shuzhen Wu
- Department of Obstetrics, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Shaoxin Ye
- Department of Fetal Medicine Research, Foshan Fetal Medicine Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
- Department of Obstetrics, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Huiting Ma
- Department of Fetal Medicine Research, Foshan Fetal Medicine Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| | - Zhengping Liu
- Department of Fetal Medicine Research, Foshan Fetal Medicine Research Institute, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
- Department of Obstetrics, Foshan Women and Children's Hospital Affiliated to Southern Medical University, Foshan, Guangdong 528000, P.R. China
| |
Collapse
|
18
|
Erben A, Hörning M, Hartmann B, Becke T, Eisler SA, Southan A, Cranz S, Hayden O, Kneidinger N, Königshoff M, Lindner M, Tovar GEM, Burgstaller G, Clausen‐Schaumann H, Sudhop S, Heymann M. Precision 3D-Printed Cell Scaffolds Mimicking Native Tissue Composition and Mechanics. Adv Healthc Mater 2020; 9:e2000918. [PMID: 33025765 DOI: 10.1002/adhm.202000918] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/29/2020] [Indexed: 12/20/2022]
Abstract
Cellular dynamics are modeled by the 3D architecture and mechanics of the extracellular matrix (ECM) and vice versa. These bidirectional cell-ECM interactions are the basis for all vital tissues, many of which have been investigated in 2D environments over the last decades. Experimental approaches to mimic in vivo cell niches in 3D with the highest biological conformity and resolution can enable new insights into these cell-ECM interactions including proliferation, differentiation, migration, and invasion assays. Here, two-photon stereolithography is adopted to print up to mm-sized high-precision 3D cell scaffolds at micrometer resolution with defined mechanical properties from protein-based resins, such as bovine serum albumin or gelatin methacryloyl. By modifying the manufacturing process including two-pass printing or post-print crosslinking, high precision scaffolds with varying Young's moduli ranging from 7-300 kPa are printed and quantified through atomic force microscopy. The impact of varying scaffold topographies on the dynamics of colonizing cells is observed using mouse myoblast cells and a 3D-lung microtissue replica colonized with primary human lung fibroblast. This approach will allow for a systematic investigation of single-cell and tissue dynamics in response to defined mechanical and bio-molecular cues and is ultimately scalable to full organs.
Collapse
Affiliation(s)
- Amelie Erben
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Marcel Hörning
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
| | - Bastian Hartmann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Tanja Becke
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stephan A. Eisler
- Stuttgart Research Center Systems Biology University of Stuttgart Nobelstr. 15 Stuttgart 70569 Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Séverine Cranz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Oliver Hayden
- Heinz‐Nixdorf‐Chair of Biomedical Electronics, TranslaTUM, Campus Klinikum rechts der Isar Technical University of Munich Einsteinstraße 25 Munich 81675 Germany
| | - Nikolaus Kneidinger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Department of Internal Medicine V Ludwig‐Maximillians‐University Munich Marchioninistr. 15 Munich 81377 Germany
| | - Melanie Königshoff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Research Unit Lung Repair and Regeneration Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University of Colorado Department of Pulmonary Sciences and Critical Care Medicine 13001 E. 17th Pl. Aurora CO 80045 USA
| | - Michael Lindner
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- University Department of Visceral and Thoracic Surgery Salzburg Paracelsus Medical University Müllner Hauptstraße 48 Salzburg A‐5020 Austria
| | - Günter E. M. Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP University of Stuttgart Nobelstr. 12 Stuttgart 70569 Germany
| | - Gerald Burgstaller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC‐M bioArchive, Helmholtz Zentrum München Member of the German Center for Lung Research (DZL) Max‐Lebsche‐Platz 31 Munich 81377 Germany
- Institute of Lung Biology and Disease (ILBD) Helmholtz Zentrum München Max‐Lebsche‐Platz 31 Munich 81377 Germany
| | - Hauke Clausen‐Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Stefanie Sudhop
- Center for Applied Tissue Engineering and Regenerative Medicine Munich University of Applied Sciences Lothstr. 34 Munich 80533 Germany
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
| | - Michael Heymann
- Center for NanoScience (CeNS) Ludwig‐Maximilians‐University Geschwister‐Scholl Platz 1 Munich 80539 Germany
- Institute of Biomaterials and Biomolecular Systems University of Stuttgart Pfaffenwaldring 57 Stuttgart 70569 Germany
- Department of Cellular and Molecular Biophysics MPI of Biochemistry Martinsried Am Klopferspitz 18 Planegg 82152 Germany
| |
Collapse
|
19
|
Kimicata M, Swamykumar P, Fisher JP. Extracellular Matrix for Small-Diameter Vascular Grafts. Tissue Eng Part A 2020; 26:1388-1401. [PMID: 33231135 PMCID: PMC7759287 DOI: 10.1089/ten.tea.2020.0201] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
To treat coronary heart disease, coronary artery bypass grafts are used to divert blood flow around blockages in the coronary arteries. Autologous grafts are the gold standard of care, but they are characterized by their lack of availability, low quality, and high failure rates. Alternatively, tissue-engineered small-diameter vascular grafts made from synthetic or natural polymers have not demonstrated adequate results to replace autologous grafts; synthetic grafts result in a loss of patency due to thrombosis and intimal hyperplasia, whereas scaffolds from natural polymers are generally unable to support the physiological conditions. Extracellular matrix (ECM) from a variety of sources, including cell-derived, 2D, and cannular tissues, has become an increasingly useful tool for this application. The current review examines the ECM-based methods that have recently been investigated in the field and comments on their viability for clinical applications.
Collapse
Affiliation(s)
- Megan Kimicata
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland, USA
- Center for Engineering Complex Tissues, and University of Maryland, College Park, Maryland, USA
| | - Prateek Swamykumar
- Center for Engineering Complex Tissues, and University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - John P. Fisher
- Center for Engineering Complex Tissues, and University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
20
|
Zambuto SG, Clancy KBH, Harley BAC. Tuning Trophoblast Motility in a Gelatin Hydrogel via Soluble Cues from the Maternal-Fetal Interface. Tissue Eng Part A 2020; 27:1064-1073. [PMID: 33216701 DOI: 10.1089/ten.tea.2020.0097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Trophoblast cells play multiple critical roles in pregnancy, notably modulating blastocyst attachment to the endometrium as well as invading into and actively remodeling the endometrium to facilitate biotransport needs of the growing embryo. Despite the importance of trophoblast invasion for processes essential at early stages of pregnancy, much remains unknown regarding the balance of signaling molecules that may influence trophoblast invasion into the endometrium. The goal of this study was to use three-dimensional trophoblast spheroid motility assays to examine the effect of cues from the maternal-fetal interface on trophoblast motility. We report use of a methacrylamide-functionalized gelatin hydrogel to support quantitative analysis of trophoblast outgrowth area and cell viability. We show that this multidimensional model of trophoblast motility can resolve quantifiable differences in outgrowth area and viability in the presence of a known invasion promoter, epidermal growth factor, and a known invasion inhibitor, transforming growth factor β1. We then investigate the sensitivity of trophoblast motility to cortisol, a hormone associated with exogenous stressors. Together, this approach provides a toolset to investigate the coordinated action of physiological and pathophysiological processes on early stages of trophoblast invasion.
Collapse
Affiliation(s)
- Samantha G Zambuto
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Kathryn B H Clancy
- Department of Anthropology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois Urbana-Champaign, Urbana, Illinois, USA.,Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
21
|
Liu M, Yin Y, Yu H, Zhou R. Laminins Regulate Placentation and Pre-eclampsia: Focus on Trophoblasts and Endothelial Cells. Front Cell Dev Biol 2020; 8:754. [PMID: 32850857 PMCID: PMC7426496 DOI: 10.3389/fcell.2020.00754] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 07/20/2020] [Indexed: 01/03/2023] Open
Abstract
Pre-eclampsia is a systemic vascular disease characterized by new-onset hypertension and/or proteinuria at ≥20 weeks of gestation and leads to high rates of maternal and perinatal morbidity and mortality. Despite the incomplete understanding of pre-eclampsia pathophysiology, it is accepted that insufficient spiral artery remodeling and endothelial dysfunction are major contributors. Laminins (LNs) are a vital family of extracellular matrix (ECM) molecules present in basement membranes that provide unique spatial and molecular information to regulate implantation and placentation. LNs interact with cell surface receptors to trigger intracellular signals that affect cellular behavior. This mini-review summarizes the role of LNs in placental development during normal pregnancy. Moreover, it describes how LN deficiency can lead to the pre-eclampsia, which is associated with trophoblast and vascular endothelial dysfunction. New research directions and the prospect of clinical diagnosis of LN deficiency are discussed, and the gaps in basic and clinical research in this field are highlighted.
Collapse
Affiliation(s)
- Min Liu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yangxue Yin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Hongbiao Yu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Rong Zhou
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University; Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
22
|
Arumugasaamy N, Rock KD, Kuo CY, Bale TL, Fisher JP. Microphysiological systems of the placental barrier. Adv Drug Deliv Rev 2020; 161-162:161-175. [PMID: 32858104 DOI: 10.1016/j.addr.2020.08.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022]
Abstract
Methods to evaluate maternal-fetal transport across the placental barrier have generally involved clinical observations after-the-fact, ex vivo perfused placenta studies, or in vitro Transwell assays. Given the ethical and technical limitations in these approaches, and the drive to understand fetal development through the lens of transport-induced injury, such as with the examples of thalidomide and Zika Virus, efforts to develop novel approaches to study these phenomena have expanded in recent years. Notably, within the past 10 years, placental barrier models have been developed using hydrogel, bioreactor, organ-on-a-chip, and bioprinting approaches. In this review, we discuss the biology of the placental barrier and endeavors to recapitulate this barrier in vitro using these approaches. We also provide analysis of current limitations to drug discovery in this context, and end with a future outlook.
Collapse
|
23
|
Advanced Fabrication Techniques of Microengineered Physiological Systems. MICROMACHINES 2020; 11:mi11080730. [PMID: 32731495 PMCID: PMC7464561 DOI: 10.3390/mi11080730] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/21/2022]
Abstract
The field of organs-on-chips (OOCs) has experienced tremendous growth over the last decade. However, the current main limiting factor for further growth lies in the fabrication techniques utilized to reproducibly create multiscale and multifunctional devices. Conventional methods of photolithography and etching remain less useful to complex geometric conditions with high precision needed to manufacture the devices, while laser-induced methods have become an alternative for higher precision engineering yet remain costly. Meanwhile, soft lithography has become the foundation upon which OOCs are fabricated and newer methods including 3D printing and injection molding show great promise to innovate the way OOCs are fabricated. This review is focused on the advantages and disadvantages associated with the commonly used fabrication techniques applied to these microengineered physiological systems (MPS) and the obstacles that remain in the way of further innovation in the field.
Collapse
|
24
|
The relationship among vitamin D, TLR4 pathway and preeclampsia. Mol Biol Rep 2020; 47:6259-6267. [PMID: 32654051 DOI: 10.1007/s11033-020-05644-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/03/2020] [Indexed: 10/23/2022]
Abstract
Preeclampsia is a pregnancy-specific syndrome that has been the greatest cause of maternal and fetal morbidity and mortality. The impaired outcomes are related to maternal and the offspring healthy in the short and long-term. Although preeclampsia origins remain unclear, it is well known that there is impaired trophoblast invasion with culminant abnormal immune response. The early and late-onset preeclampsia have been studied, the subtypes have the same difference in the placentation and inflammatory features. Dietary compounds can stimulate or inhibit the activation of immune cells. Low vitamin D intake has been linked to impaired fetal development, intrauterine growth restriction, and preeclampsia. Vitamin D has been described as an anti-inflammatory effect. It can downregulate pro-inflammatory cytokines expression by the inhibition of the Nuclear Factor-ĸB pathway signaling cascade. High vitamin D levels could attenuate the immune response. On the other hand, vitamin D deficiency may contribute to increasing pro-inflammatory state. In preeclampsia, there is a reduced expression of vitamin D receptor and its metabolism is disrupted. In this review, we aimed to discuss the role of vitamin D as an anti-inflammatory agent in relation to the pro-inflammatory process of preeclampsia through the activation of the TLR4 pathway. Although there are limited studies showing the relation between vitamin D and lower risk of preeclampsia, the maternal status of vitamin D seems to influence the risk of PE development. Therefore, vitamin D supplementation in women may be a strategy to improve pregnancy outcomes.
Collapse
|
25
|
Cui J, Chen X, Lin S, Li L, Fan J, Hou H, Li P. MiR-101-containing extracellular vesicles bind to BRD4 and enhance proliferation and migration of trophoblasts in preeclampsia. Stem Cell Res Ther 2020; 11:231. [PMID: 32527308 PMCID: PMC7291671 DOI: 10.1186/s13287-020-01720-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/24/2020] [Accepted: 05/08/2020] [Indexed: 02/06/2023] Open
Abstract
Background Preeclampsia (PE) is a frequently occurring pregnancy disorder in the placenta, which results in various maternal and fetal complications. The current study aims to evaluate the role of extracellular vesicles (EVs)-encapsulated microRNA (miR)-101 in biological processes of trophoblasts in PE and its underlying mechanism. Methods Human umbilical cord mesenchymal stem cell (HUCMSC) and HUCMSC-derived EVs were isolated and cultured, after which EV characterization was carried out using PKH67 staining. In silico analyses were adopted to predict the downstream target genes of miR-101, and dual luciferase reporter gene assay was applied to validate the binding affinity. Furthermore, loss- and gain-of-function approaches were adopted to determine the role of miR-101 and bromodomain-containing protein 4 (BRD4) in trophoblast proliferation and invasion using EDU staining and transwell assay. In addition, a rat model of PE was established to verify the function of EV-encapsulated miR-101 in vivo. Results Placental tissues obtained from PE patients presented with downregulated miR-101 expression and upregulated BRD4 and CXCL11 expression. EV-encapsulated miR-101 from HUCMSCs could be delivered into the trophoblast HTR-8/SVneo cells, thus enhancing proliferation and migration of trophoblasts. Mechanically, miR-101 targeted and negatively regulated BRD4 expression. BRD4 knockdown promoted the proliferation and migration of trophoblasts by suppressing NF-κB/CXCL11 axis. EV-encapsulated miR-101 from HUCMSCs also reduced blood pressure and 24 h urine protein in vivo, thereby ameliorating PE. Conclusion In summary, EV-encapsulated miR-101 promoted proliferation and migration of placental trophoblasts through the inhibition of BRD4 expression via NF-κB/CXCL11 inactivation.
Collapse
Affiliation(s)
- Jinhui Cui
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China
| | - Xinjuan Chen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China
| | - Shuo Lin
- Department of Endocrinology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 516000, People's Republic of China
| | - Ling Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China
| | - Jianhui Fan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China
| | - Hongying Hou
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China
| | - Ping Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Sun Yat-Sen University, No. 600, Tianhe Road, Guangzhou, 516000, Guangdong Province, People's Republic of China.
| |
Collapse
|
26
|
Tang SW, Tong WY, Pang SW, Voelcker NH, Lam YW. Deconstructing, Replicating, and Engineering Tissue Microenvironment for Stem Cell Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:540-554. [PMID: 32242476 DOI: 10.1089/ten.teb.2020.0044] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
One of the most crucial components of regenerative medicine is the controlled differentiation of embryonic or adult stem cells into the desired cell lineage. Although most of the reported protocols of stem cell differentiation involve the use of soluble growth factors, it is increasingly evident that stem cells also undergo differentiation when cultured in the appropriate microenvironment. When cultured in decellularized tissues, for instance, stem cells can recapitulate the morphogenesis and functional specialization of differentiated cell types with speed and efficiency that often surpass the traditional growth factor-driven protocols. This suggests that the tissue microenvironment (TME) provides stem cells with a holistic "instructive niche" that harbors signals for cellular reprogramming. The translation of this into medical applications requires the decoding of these signals, but this has been hampered by the complexity of TME. This problem is often addressed by a reductionist approach, in which cells are exposed to substrates decorated with simple, empirically designed geometries, textures, and chemical compositions ("bottom-up" approach). Although these studies are invaluable in revealing the basic principles of mechanotransduction mechanisms, their physiological relevance is often uncertain. This review examines the recent progress of an alternative, "top-down" approach, in which the TME is treated as a holistic biological entity. This approach is made possible by recent advances in systems biology and fabrication technologies that enable the isolation, characterization, and reconstitution of TME. It is hoped that these new techniques will elucidate the nature of niche signals so that they can be extracted, replicated, and controlled. This review summarizes these emerging techniques and how the data they generated are changing our view on TME. Impact statement This review summarizes the current state of art of the understanding of instructive niche in the field of tissue microenvironment. Not only did we survey the use of different biochemical preparations as stimuli of stem cell differentiation and summarize the recent effort in dissecting the biochemical composition of these preparations, through the application of extracellular matrix (ECM) arrays and proteomics, but we also introduce the use of open-source, high-content immunohistochemistry projects in contributing to the understanding of tissue-specific composition of ECM. We believe this review would be highly useful for our peer researching in the same field. "Mr. Tulkinghorn is always the same… so oddly out of place and yet so perfectly at home." -Charles Dickens, Bleak House.
Collapse
Affiliation(s)
- Sze Wing Tang
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| | - Wing Yin Tong
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Stella W Pang
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong, Hong Kong
| | - Nicolas H Voelcker
- Melbourne Center for Nanofabrication, Victorian Node of the Australian National Fabrication, Clayton, Australia.,Drug Delivery Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Yun Wah Lam
- Department of Chemistry, City University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
27
|
Yu JR, Janssen M, Liang BJ, Huang HC, Fisher JP. A liposome/gelatin methacrylate nanocomposite hydrogel system for delivery of stromal cell-derived factor-1α and stimulation of cell migration. Acta Biomater 2020; 108:67-76. [PMID: 32194261 PMCID: PMC7198368 DOI: 10.1016/j.actbio.2020.03.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 03/09/2020] [Accepted: 03/11/2020] [Indexed: 11/29/2022]
Abstract
Chronic, non-healing skin and soft tissue wounds are susceptible to infection, difficult to treat clinically, and can severely reduce a patient's quality of life. A key aspect of this issue is the impaired recruitment of mesenchymal stem cells (MSCs), which secrete regenerative cytokines and modulate the phenotypes of other effector cells that promote healing. We have engineered a therapeutic delivery system that can controllably release the pro-healing chemokine stromal cell derived factor-1α (SDF-1α) to induce the migration of MSCs. In order to protect the protein cargo from hydrolytic degradation and control its release, we have loaded SDF-1α in anionic liposomes (lipoSDF) and embedded them in gelatin methacrylate (GelMA) to form a nanocomposite hydrogel. In this study, we quantify the release of SDF-1α from our hydrogel system and measure the induced migration of MSCs in vitro via a transwell assay. Lastly, we evaluate the ability of this system to activate intracellular signaling in MSCs by using Western blots to probe for the phosphorylation of key proteins in the mTOR pathway. To our knowledge, this is the first study to report the delivery of liposomal SDF-1α using a nanocomposite approach. The results of this study expand on our current understanding of factors that can be modified to affect MSC behavior and phenotype. Furthermore, our findings contribute to the development of new hydrogel-based therapeutic delivery strategies for clinical wound healing applications. STATEMENT OF SIGNIFICANCE: Chronic, non-healing wounds promote an inflammatory environment that inhibits the migration of mesenchymal stem cells (MSCs), which secrete pro-healing and regenerative cytokines. The goal of this project is to apply principles of tissue engineering to achieve controllable release of the pro-healing chemokine SDF-1α to modulate the intracellular signaling and migratory behavior of MSCs. In this work, we introduce a nanocomposite strategy to tailor the release of SDF-1α using a liposome/gelatin methacrylate hydrogel approach. We are the first group to report the delivery of liposomal SDF-1α using this strategy. Our findings aim to further elucidate the role of MSCs in directing wound healing and guide the development of immunomodulatory and therapeutic delivery strategies for clinical wound healing applications.
Collapse
Affiliation(s)
- Justine R Yu
- Fischell Department of Bioengineering, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States; NIH/NBIB Center for Engineering Complex Tissues, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States; University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Miriam Janssen
- Fischell Department of Bioengineering, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States
| | - Barry J Liang
- Fischell Department of Bioengineering, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States
| | - Huang-Chiao Huang
- Fischell Department of Bioengineering, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States; Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States; NIH/NBIB Center for Engineering Complex Tissues, University of Maryland - College Park, 3121 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742, United States.
| |
Collapse
|
28
|
Abstract
Advances in stem cell cultures and human-induced pluripotent stem cells have inculcated interests in a rapidly evolving concept – ”organoids.” These are three-dimensional (3D) structures mimicking some of the phenomena of the real organs at anatomical, multicellular, and functional levels in vitro. Organoids have been proven to be better than two-dimensional cell culture in replicating the functionality, architectural, and geometrical features of tissues in vivo. Recent advancements have led to the generation of models for organ development and disease, finding applications in the drug discovery, screening of novel compounds, and personalized medicine. Since organoids follow the same natural pathway as the normal tissue or pathology, they can be used to study the expression of various genotypes and phenotypic variations across different species. In the light of these advancements, organoids are now being merged with bioengineering to come up with even better and reliable models to predict the disease progression and effectiveness of precision medicines, few of its important applications. This article discusses the various aspects of this emerging concept along with its uses, both in the present times and near future, with a special focus on pharmacological applications.
Collapse
Affiliation(s)
| | - Shubham Atal
- Department of Pharmacology, AIIMS, Bhopal, Madhya Pradesh, India
| | - Avik Ray
- Department of Pharmacology, AIIMS, Bhopal, Madhya Pradesh, India
| | - C A Pravin
- Department of Pharmacology, AIIMS, Bhopal, Madhya Pradesh, India
| | - Malaya Nanda
- Department of Pharmacology, AIIMS, Bhopal, Madhya Pradesh, India
| |
Collapse
|
29
|
Belleghem SMV, Mahadik B, Snodderly KL, Fisher JP. Overview of Tissue Engineering Concepts and Applications. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00081-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
30
|
Ding H, Illsley NP, Chang RC. 3D Bioprinted GelMA Based Models for the Study of Trophoblast Cell Invasion. Sci Rep 2019; 9:18854. [PMID: 31827129 PMCID: PMC6906490 DOI: 10.1038/s41598-019-55052-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 11/12/2019] [Indexed: 12/21/2022] Open
Abstract
Bioprinting is an emerging and promising technique for fabricating 3D cell-laden constructs for various biomedical applications. In this paper, we employed 3D bioprinted GelMA-based models to investigate the trophoblast cell invasion phenomenon, enabling studies of key placental functions. Initially, a set of optimized material and process parameters including GelMA concentration, UV crosslinking time and printing configuration were identified by systematic, parametric study. Following this, a multiple-ring model (2D multi-ring model) was tested with the HTR-8/SVneo trophoblast cell line to measure cell movement under the influence of EGF (chemoattractant) gradients. In the multi-ring model, the cell front used as a cell invasion indicator moves at a rate of 85 ± 33 µm/day with an EGF gradient of 16 µM. However, the rate was dramatically reduced to 13 ± 5 µm/day, when the multi-ring model was covered with a GelMA layer to constrain cells within the 3D environment (3D multi-ring model). Due to the geometric and the functional limitations of multi-ring model, a multi-strip model (2D multi-strip model) was developed to investigate cell movement in the presence and absence of the EGF chemoattractant. The results show that in the absence of an overlying cell-free layer of GelMA, movement of the cell front shows no significant differences between control and EGF-stimulated rates, due to the combination of migration and proliferation at high cell density (6 × 106 cells/ml) near the GelMA surface. When the model was covered by a layer of GelMA (3D multi-strip model) and migration was excluded, EGF-stimulated cells showed an invasion rate of 21 ± 3 µm/day compared to the rate for unstimulated cells, of 5 ± 4 µm/day. The novel features described in this report advance the use of the 3D bioprinted placental model as a practical tool for not only measurement of trophoblast invasion but also the interaction of invading cells with other tissue elements.
Collapse
Affiliation(s)
- Houzhu Ding
- Stevens Institute of Technology, Department of Mechanical Engineering, Hoboken, NJ, 07030, USA
| | - Nicholas P Illsley
- Hackensack University Medical Center, Department of Obstetrics and Gynecology, Hackensack, NJ, 07601, USA
| | - Robert C Chang
- Stevens Institute of Technology, Department of Mechanical Engineering, Hoboken, NJ, 07030, USA.
| |
Collapse
|
31
|
Wu S, Zhang H, Liu Y, Wang R, Ye S, Zeng M, Liu Z. [Long non-coding RNAs show different expression profiles and display competing endogenous RNA potential in placenta accreta spectrum disorders]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:1253-1259. [PMID: 31801721 DOI: 10.12122/j.issn.1673-4254.2019.10.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the expression profile of long non-coding RNAs (lncRNA) and identify potential lncRNA-related competing endogenous RNAs (ceRNA) in placenta accrete spectrum disorders (PAS). METHODS Five tissue specimens of placental implantation and 5 adjacent normal placental tissues were collected from cesarean section deliveries complicated by PAS in our hospital between December, 2017 and June, 2018. Human microarrays were used to identify the lncRNAs that were differentially expressed in PAS, and 5 of the identified lncRNAs were further validated using qRT-PCR. GO and KEGG pathway analyses were performed to indentify the most significant enrichment functions. A ceRNA network was constructed based on ENST00000511361 (RP5-875H18.4), NR_027457 (LINC00221) and NR_126415 (FOXP4-AS1) to pinpoint the potential lncRNAs-related ceRNA. RESULTS A total of 329 lncRNAs and 179 mRNAs were identified to have differential expression in PAS. The results of qRT-PCR were consistent with the human microarrays results. Transforming growth factor-β (TGF-β) signaling pathway was the most significantly enriched pathway. The constructed ceRNA network suggested that RP5-875H18.4--miRNA-218--SLIT2 had a potential ceRNA regulatory mechanism in PAS. CONCLUSIONS The differentially expressed lncRNAs are involved in the occurrence and progression of PAS possibly by regulating the TGF-β signaling pathway. The ceRNA network of RP5-875H18.4--miRNA-218--SLIT2 may play a role in the occurrence of PAS.
Collapse
Affiliation(s)
- Shuzhen Wu
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - Huishan Zhang
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China.,Foshan Fetal Medicine Institute, Foshan 528000, China
| | - Yan Liu
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - Rui Wang
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - Shaoxin Ye
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China.,Foshan Fetal Medicine Institute, Foshan 528000, China
| | - Meng Zeng
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China
| | - Zhengping Liu
- Department of Obstetrics, Foshan Maternal and Child Health Hospital Affiliated to Southern Medical University, Foshan 528000, China.,Foshan Fetal Medicine Institute, Foshan 528000, China
| |
Collapse
|
32
|
Arumugasaamy N, Hurley-Novatny A, Lembong J, Kim PC, Fisher JP. Assessing SSRIs' effects on fetal cardiomyocytes utilizing placenta-fetus model. Acta Biomater 2019; 99:258-268. [PMID: 31536839 DOI: 10.1016/j.actbio.2019.09.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 09/09/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022]
Abstract
Selective serotonin reuptake inhibitors (SSRIs) have been shown to hinder cardiomyocyte signaling, raising concerns about their safety during pregnancy. Approaches to assess SSRI-induced effects on fetal cardiovascular cells following passage of drugs through the placental barrier in vitro have only recently become available. Herein, we report that the SSRIs, fluoxetine and sertraline, lead to slowed cardiomyocyte calcium oscillations and induce increased secretion of troponin T and creatine kinase-MB with reduced secretion of NT-proBNP, three key cardiac injury biomarkers. We show the cardiomyocyte calcium handling effects are further amplified following indirect exposure through a placental barrier model. These studies are the first to investigate the effects of placental barrier co-culture with cardiomyocytes in vitro and to show cardiotoxicity of SSRIs following passage through the placental barrier. STATEMENT OF SIGNIFICANCE: Use of selective serotonin reuptake inhibitors (SSRIs), a class of antidepressants, during pregnancy continues to rise despite multiple studies showing potential for detrimental effects on the developing fetus. SSRIs are particularly thought to slow cardiovascular electrical activity, such as ion signaling, yet few, if any, methods exist to rigorously study these drug-induced effects on human pregnancy and the developing fetus. Within this study, we utilized a placenta-fetus model to evaluate these drug-induced effects on cardiomyocytes, looking the drugs' effects on calcium handling and secretion of multiple cardiac injury biomarkers. Together, with existing literature, this study provides a platform for assessing pharmacologic effects of drugs on cells mimicking the fetus and the role the placenta plays in this process.
Collapse
|
33
|
Arumugasaamy N, Gudelsky A, Hurley-Novatny A, Kim PC, Fisher JP. Model Placental Barrier Phenotypic Response to Fluoxetine and Sertraline: A Comparative Study. Adv Healthc Mater 2019; 8:e1900476. [PMID: 31407872 PMCID: PMC6752965 DOI: 10.1002/adhm.201900476] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/10/2019] [Indexed: 12/20/2022]
Abstract
Medications taken during pregnancy may significantly impact fetal development, yet there are few studies that rigorously assess medication safety due to ethical concerns. Selective serotonin reuptake inhibitors (SSRIs) are a class of drug increasingly being prescribed for depression, yet multiple studies have shown that taking SSRIs during pregnancy can lead to preterm birth and potential health concerns for the baby. Therefore, a biomimetic placental barrier model is utilized herein to assess transport profiles and phenotypic effects resulting from SSRI exposure, comparing fluoxetine and sertraline. Results show that the placental barrier quickly uptakes drug from the maternal side, but slowly releases on the fetal side. Phenotypically, there is a dose-dependent change in cell adhesion molecule (CAM) and transforming growth factor beta (TGFβ) secretions, markers of cell adhesion and angiogenesis. Both drugs impact CAM secretions, whereas sertraline alone impacts TGFβ secretions. When evaluating cell type, it becomes clear that endothelial cells, not trophoblast, are the main cell type involved in these phenotypic changes. Overall, these findings further the understanding of SSRI transplacental transport and drug-induced effects on the placental barrier.
Collapse
Affiliation(s)
- Navein Arumugasaamy
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Alana Gudelsky
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
| | - Amelia Hurley-Novatny
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| | - Peter C.W. Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, D.C. 20010
- Department of Surgery, The George Washington University, Washington, D.C. 20037
| | - John P. Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD 20742
| |
Collapse
|
34
|
Lerman MJ, Muramoto S, Arumugasaamy N, Van Order M, Lembong J, Gerald AG, Gillen G, Fisher JP. Development of surface functionalization strategies for 3D-printed polystyrene constructs. J Biomed Mater Res B Appl Biomater 2019; 107:2566-2578. [PMID: 30821930 DOI: 10.1002/jbm.b.34347] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 01/18/2019] [Accepted: 02/10/2019] [Indexed: 01/17/2023]
Abstract
There is a growing interest in 3D printing to fabricate culture substrates; however, the surface properties of the scaffold remain pertinent to elicit targeted and expected cell responses. Traditional 2D polystyrene (PS) culture systems typically require surface functionalization (oxidation) to facilitate and encourage cell adhesion. Determining the surface properties which enhance protein adhesion from media and cellular extracellular matrix (ECM) production remains the first step to translating 2D PS systems to a 3D culture surface. Here we show that the presence of carbonyl groups to PS surfaces correlated well with successful adhesion of ECM proteins and sustaining ECM production of deposited human mesenchymal stem cells, if the surface has a water contact angle between 50° and 55°. Translation of these findings to custom-fabricated 3D PS scaffolds reveals carbonyl groups continued to enhance spreading and growth in 3D culture. Cumulatively, these data present a method for 3D printing PS and the design considerations required for understanding cell-material interactions. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B:2566-2578, 2019.
Collapse
Affiliation(s)
- Max J Lerman
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland.,Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland.,Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland
| | - Shin Muramoto
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - Navein Arumugasaamy
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland.,Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Health System, Washington, District of Columbia
| | - Michael Van Order
- Department of Materials Science and Engineering, University of Maryland, College Park, Maryland
| | - Josephine Lembong
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Anushka G Gerald
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| | - Greg Gillen
- Surface and Trace Chemical Analysis Group, Materials Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland
| | - John P Fisher
- Center for Engineering Complex Tissues, University of Maryland, College Park, Maryland.,Fischell Department of Bioengineerin, University of Maryland, College Park, Maryland
| |
Collapse
|
35
|
DaSilva-Arnold SC, Kuo CY, Davra V, Remache Y, Kim PCW, Fisher JP, Zamudio S, Al-Khan A, Birge RB, Illsley NP. ZEB2, a master regulator of the epithelial-mesenchymal transition, mediates trophoblast differentiation. Mol Hum Reprod 2019; 25:61-75. [PMID: 30462321 PMCID: PMC6497037 DOI: 10.1093/molehr/gay053] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/09/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022] Open
Abstract
STUDY QUESTION Does the upregulation of the zinc finger E-box binding homeobox 2 (ZEB2) transcription factor in human trophoblast cells lead to alterations in gene expression consistent with an epithelial-mesenchymal transition (EMT) and a consequent increase in invasiveness? SUMMARY ANSWER Overexpression of ZEB2 results in an epithelial-mesenchymal shift in gene expression accompanied by a substantial increase in the invasive capacity of human trophoblast cells. WHAT IS KNOWN ALREADY In-vivo results have shown that cytotrophoblast differentiation into extravillous trophoblast involves an epithelial-mesenchymal transition. The only EMT master regulatory factor which shows changes consistent with extravillous trophoblast EMT status and invasive capacity is the ZEB2 transcription factor. STUDY DESIGN, SIZE, DURATION This study is a mechanistic investigation of the role of ZEB2 in trophoblast differentiation. We generated stable ZEB2 overexpression clones using the epithelial BeWo and JEG3 choriocarcinoma lines. Using these clones, we investigated the effects of ZEB2 overexpression on the expression of EMT-associated genes and proteins, cell morphology and invasive capability. PARTICIPANTS/MATERIALS, SETTING, METHODS We used lentiviral transduction to overexpress ZEB2 in BeWo and JEG3 cells. Stable clones were selected based on ZEB2 expression and morphology. A PCR array of EMT-associated genes was used to probe gene expression. Protein measurements were performed by western blotting. Gain-of-function was assessed by quantitatively measuring cell invasion rates using a Transwell assay, a 3D bioprinted placenta model and the xCelligenceTM platform. MAIN RESULTS AND THE ROLE OF CHANCE The four selected clones (2 × BeWo, 2 × JEG3, based on ZEB2 expression and morphology) all showed gene expression changes indicative of an EMT. The two clones (1 × BeWo, 1 × JEG3) showing >40-fold increase in ZEB2 expression also displayed increased ZEB2 protein; the others, with increases in ZEB2 expression <14-fold did not. The two high ZEB2-expressing clones demonstrated robust increases in invasive capacity, as assessed by three types of invasion assay. These data identify ZEB2-mediated transcription as a key mechanism transforming the epithelial-like trophoblast into cells with a mesenchymal, invasive phenotype. LARGE SCALE DATA PCR array data have been deposited in the GEO database under accession number GSE116532. LIMITATIONS, REASONS FOR CAUTION These are in-vitro studies using choriocarcinoma cells and so the results should be interpreted in view of these limitations. Nevertheless, the data are consistent with in-vivo findings and are replicated in two different cell lines. WIDER IMPLICATIONS OF THE FINDINGS The combination of these data with the in-vivo findings clearly identify ZEB2-mediated EMT as the mechanism for cytotrophoblast differentiation into extravillous trophoblast. Having characterized these cellular mechanisms, it will now be possible to identify the intracellular and extracellular regulatory components which control ZEB2 and trophoblast differentiation. It will also be possible to identify the aberrant factors which alter differentiation in invasive pathologies such as preeclampsia and abnormally invasive placenta (AKA accreta, increta, percreta). STUDY FUNDING AND COMPETING INTEREST(s) Funding was provided by the Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Surgery at Hackensack Meridian Health, Hackensack, NJ. The 3D bioprinted placental model work done in Drs Kim and Fisher's labs was supported by the Children's National Medical Center. The xCELLigence work done in Dr Birge's lab was supported by NIH CA165077. The authors declare no competing interests.
Collapse
Affiliation(s)
- Sonia C DaSilva-Arnold
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery and Center for Abnormal Placentation, Hackensack University Medical Center, Hackensack, NJ, USA
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Che-Ying Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD, USA
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington DC, USA
| | - Viralkumar Davra
- Department of Microbiology, Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Yvonne Remache
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery and Center for Abnormal Placentation, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Peter C W Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington DC, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
- NIH Center for Engineering Complex Tissues, University of Maryland, College Park, MD, USA
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington DC, USA
| | - Stacy Zamudio
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery and Center for Abnormal Placentation, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Abdulla Al-Khan
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery and Center for Abnormal Placentation, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Raymond B Birge
- Department of Microbiology, Biochemistry and Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Nicholas P Illsley
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine and Surgery and Center for Abnormal Placentation, Hackensack University Medical Center, Hackensack, NJ, USA
| |
Collapse
|
36
|
Kuo CY, Shevchuk M, Opfermann J, Guo T, Santoro M, Fisher JP, Kim PCW. Trophoblast-endothelium signaling involves angiogenesis and apoptosis in a dynamic bioprinted placenta model. Biotechnol Bioeng 2019; 116:181-192. [PMID: 30298908 PMCID: PMC6289739 DOI: 10.1002/bit.26850] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 09/26/2018] [Accepted: 10/04/2018] [Indexed: 01/01/2023]
Abstract
Trophoblast invasion and remodeling of the maternal spiral arteries are required for pregnancy success. Aberrant endothelium-trophoblast crosstalk may lead to preeclampsia, a pregnancy complication that has serious effects on both the mother and the baby. However, our understanding of the mechanisms involved in this pathology remains elementary because the current in vitro models cannot describe trophoblast-endothelium interactions under dynamic culture. In this study, we developed a dynamic three-dimensional (3D) placenta model by bioprinting trophoblasts and an endothelialized lumen in a perfusion bioreactor. We found the 3D printed perfusion bioreactor system significantly augmented responses of endothelial cells by encouraging network formations and expressions of angiogenic markers, cluster of differentiation 31 (CD31), matrix metalloproteinase-2 (MMP2), matrix metalloproteinase-9 (MMP9), and vascular endothelial growth factor A (VEGFA). Bioprinting favored colocalization of trophoblasts with endothelial cells, similar to in vivo observations. Additional analysis revealed that trophoblasts reduced the angiogenic responses by reducing network formation and motility rates while inducing apoptosis of endothelial cells. Moreover, the presence of endothelial cells appeared to inhibit trophoblast invasion rates. These results clearly demonstrated the utility and potential of bioprinting and perfusion bioreactor system to model trophoblast-endothelium interactions in vitro. Our bioprinted placenta model represents a crucial step to develop advanced research approach that will expand our understanding and treatment options of preeclampsia and other pregnancy-related pathologies.
Collapse
Affiliation(s)
- Che-Ying Kuo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Mariya Shevchuk
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Justin Opfermann
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
| | - Ting Guo
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Marco Santoro
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, College Park, MD
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- Center for Engineering Complex Tissues, University of Maryland, College Park, MD
| | - Peter CW Kim
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Children’s National Health System, Washington, DC
- School of Medicine and Health Sciences, The George Washington University, Washington, DC
| |
Collapse
|
37
|
Mittal R, Woo FW, Castro CS, Cohen MA, Karanxha J, Mittal J, Chhibber T, Jhaveri VM. Organ‐on‐chip models: Implications in drug discovery and clinical applications. J Cell Physiol 2018; 234:8352-8380. [DOI: 10.1002/jcp.27729] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Frank W. Woo
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Carlo S. Castro
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Madeline A. Cohen
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Joana Karanxha
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Jeenu Mittal
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| | - Tanya Chhibber
- University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Studies, Panjab University Chandigarh India
| | - Vasanti M. Jhaveri
- Department of Otolaryngology University of Miami Miller School of Medicine Miami Florida
| |
Collapse
|
38
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|