Abstract
The female reproductive tract is a highly complex physiological system that consists of the ovaries, fallopian tubes, uterus, cervix, and vagina. An enhanced understanding of the molecular, cellular, and genetic mechanisms of the tract will allow for the development of more effective assisted reproductive technologies, therapeutics, and screening strategies for female specific disorders. Traditional 2-dimensional and 3-dimensional static culture systems may not always reflect the cellular and physical contexts or physicochemical microenvironment necessary to understand the dynamic exchange that is crucial for the functioning of the reproductive system. Microfluidic systems present a unique opportunity to study the female reproductive tract, as these systems recapitulate the multicellular architecture, contacts between different tissues, and microenvironmental cues that largely influence cell structure, function, behavior, and growth. This review discusses examples, challenges, and benefits of using microfluidic systems to model ovaries, fallopian tubes, endometrium, and placenta. Additionally, this review also briefly discusses the use of these systems in studying the effects of endocrine disrupting chemicals and diseases such as ovarian cancer, preeclampsia, and polycystic ovarian syndrome.
Keywords: microfluidic systems, organ-on-a-chip, female reproductive tract
The female reproductive tract is a highly complex physiological system that enables pregnancy and sustains the fetus throughout the pregnancy. The reproductive tract consists of the ovaries, fallopian tubes, uterus, cervix, and vagina. These organs have a strong interdependence and communicate via paracrine and endocrine signals at a spatial–temporal level, which is necessary to achieve the reproductive system’s functions (1-3). Primary functions of the system include maturing the ova (or the egg cells), providing an environment to allow fertilization, and supporting the developing embryo. Additionally, the system also influences biological function in the endocrine, immune, cardiovascular, and skeletal systems through the production of steroid hormones (4-8).
The health and regulation of the female reproductive tract is vulnerable to genetic susceptibilities, environmental factors, and off-target effects from various drugs which can result in reproductive health concerns such as polycystic ovarian syndrome, ovarian cancer, and endometriosis (9-13). The Centers for Disease Control (CDC) reported that between 2015 and 2017, about 13.1% of women from 15 to 49 years of age had impaired fecundity, and 8.8% of married women in the same age group were infertile (14). During the same period, 12.7% of women opted for fertility treatments. Predictably, intense research has gone into assisted reproductive technologies (ART), and more than 5 million children have been conceived using such technologies (15). Traditionally, this research has largely focused on increasing the likelihood of continuing pregnancy, but recently has expanded into aspects that can influence the long-term health of the fetus, such as embryonic developmental competence, gene expression patterns, and epigenetic reprogramming (16-21). The key to development of more effective ART, therapeutics, and screening strategies for female specific disorders is better understanding of the molecular, cellular, and genetic mechanisms of the female reproductive tract. However, this has been challenging due to the lack of ideal model systems.
Studying human female reproductive biology can result in some ethical concerns, especially regarding pregnant subjects, designer embryos, and contraception. Additionally, the dynamic hormonal secretion patterns and anatomical inaccessibility of the system has made it difficult to systematically study causes of infertility and the early-stage events of disorders in the female reproductive tract. Thus, 2-dimensional static culture systems have traditionally been used to study different organs and cell populations of the tract where cells are grown on tissue culture treated plastic flasks (22, 23). These allow for convenient and effective cell-based assays that help elucidate mechanisms of diseases and drugs (24). Three-dimensional (3D) culture systems, such as organoids, have also been essential tools in studying reproductive biology (25-28). These systems use synthetic polymers such as hydrogels or natural extracellular matrix molecules and can better replicate certain characteristics of the in vivo tissue architecture and signaling (29, 30). However, these systems may not always reflect the cellular and physical contexts necessary to understand the dynamic hormonal exchange that is crucial for the functioning of the human female reproductive system (31-35).
Microfluidic culture systems are a technology that has the potential to provide a more physiologically relevant model that overcomes limitations of traditional static cultures. Known by various names such as organ-on-a-chip or lab-on-a-chip, these devices can recapitulate the multicellular architecture and contacts between different tissues and thus, can enhance our understanding of in vivo mechanisms (36-38). Some of these devices offer precise control over fluid flow in a series of micrometer-sized channels, thus replicating the in vivo continuous or pulsatile perfusion (39-43). Cells and tissues in culture are exposed to mechanical cues, including fluid shear stress, tension, and compression, as well as chemical cues, such as spatiotemporally controlled gradients of secreted factors and steroid hormones (44-47). Microenvironmental cues largely influence cell structure, function, behavior, and growth (43, 48). Hence, the cellular response to externally introduced stimuli such as drugs and toxins can be more accurately represented in a microfluidic culture system (49-52).
Different organs such as the liver, kidney, lungs, and intestines have been modeled by adapting commercially available platforms such as the Emulate or Nortis chips (53-62). Instead, the reproductive field has evolved to develop unique systems that are tailored to support specific biological properties of different reproductive tissues such as elimination of polydimethylsiloxane (PDMS) to ensure availability of hydrophobic steroids, large tissue compartments for 3D follicle expansion, and recirculation of perfusion to ensure endocrine signaling (Fig. 1). Hence, this review is organized around female reproductive tract organs, including the ovaries, fallopian tubes, endometrium, and placenta, and discusses the setup of specific devices that were used to model the organ. The review also briefly discusses the use of these systems in studying effects of endocrine disrupting chemicals (EDCs) and diseases of the reproductive tract such as ovarian cancer and preeclampsia (Fig. 2). Application of microfluidic systems for the study of male reproductive system is beyond the scope of this review but has been extensively discussed in other review articles (63-68).
Figure 1.
Technical features of microfluidic systems that enable their application to the reproductive tract. Polydimethylsiloxane (PDMS) and polysulfone (PSF) are popular materials used in the fabrication of microfluidic devices. PDMS has a high tendency to absorb hydrophobic compounds, such as steroid hormones and drugs, which would impact reproductive function. Organs with a lumen benefit from an air-liquid interface to remain viable in culture but research on placental drug transport instead requires a closed fluid environment—both of these environments can be created in a microfluidic system. Unlike static culture systems, microfluidic systems can incorporate dynamic fluid flow and replicate physiochemical cues. Devices can be configured to recirculate media and reproduce endocrine loops or model circadian rhythm.
Figure 2.
Applications of microfluidics systems in studying the reproductive tract. Culturing tissues or cell populations from the female reproductive tract using microfluidic systems allows for the study of various biological processes such as ovulation, folliculogenesis, hormone biosynthesis, decidualization, postimplantation developmental events, and beating of cilia present on the fallopian tube epithelium (FTE). Microfluidic devices can assist in the study of enhanced assisted reproductive technologies (ART). Moreover, the systems can also help elucidate mechanisms, identify biomarkers, and develop therapeutic strategies for disorders of the female reproductive tract, such as high-grade serous ovarian cancer (HGSC). Microfluidic systems also provide a system for drug efficacy and toxicity, especially when used for co-culturing the tract’s organs with organs from other organ systems such as the liver and intestine.
Microfluidic Modeling of the Ovary
The ovary is the female gonad and is located near the lateral walls of the pelvic cavity. In addition to being the site of gametogenesis, the ovaries secrete steroid hormones, such as estrogen and progesterone, both of which modify the uterus and are crucial to normal reproductive development and fertility (69). The follicular phase, or the first half of the 28-day menstrual cycle, involves stimulation of ovarian follicles in response to follicle-stimulating hormone. Midway through the cycle, a spike in luteinizing hormone released by the pituitary gland triggers ovulation, an event where the preovulatory follicle ruptures and releases the oocyte. After ovulation, in the luteal phase, granulosa cells that surround the ruptured follicle undergo a process of luteinization which involves differentiation, cell enlargement, and increased progesterone production. A woman has a fixed number of oocytes at birth, unlike the sperm in males, and as a woman ages, the number of oocytes declines, resulting in menopause (70). Hence, microfluidic technology to rescue and mature the ovulated oocyte (known as the egg) might provide a route to preserving fertility, which would be particularly beneficial when treatments (such as cancer therapies) that are toxic to the arrested ovarian follicle reserve risk causing permanent infertility (17, 71, 72). Additionally, a microfluidic device that can model ovulation provides a system to study signaling pathways associated with infertility resulting from disease or compounds such as specific chemotherapy regimens (73-76).
Xiao et al (2017) designed the single and dual unit microfluidic systems, Solo-MFP and Duet-MFP, using an intermediate thermoplastic track-etched membrane (77). These devices were able to sustain murine ovarian explants, which contained follicles from all developmental stages, for the length of the human menstrual cycle. The device used pneumatic actuation technology to incorporate dynamic flow of media in their culture system and support murine follicle growth. By controlling exogenous follicle-stimulating hormone and luteinizing hormone, the group replicated maturation, ovulation, and granulosa cell luteinization. Moreover, they found that the steroid hormone profile produced by the cultured follicles mimicked that of an ovary in a human menstrual cycle. The inability of static culture to maintain proper nutrient and oxygen diffusion had previously made this challenging (78, 79).
Despite the huge potential for fertility preservation, it has been challenging to culture in vitro oocytes from immature ovarian follicles of large mammalian species because of their complex metabolic needs and long-term culture requirements (80-83). While it has previously been observed that both murine and human primary follicles behave similarly in static culture, it is essential to translate microfluidic development to larger mammalian species because of differences between mice and large mammals such as humans (84-92). For example, the menstrual cycle duration as well as follicle development period is longer in larger animals compared to mice in addition to the varied size of follicles between species (80, 93, 94). Hence, in vitro culturing of large species follicles requires a more complex and dynamic system than most static cultures can provide.
To create a dynamic system that is capable of improving survival and growth of follicles from large mammalian species in vitro, Nagashima et al (2017) built a microfluidic chip using polymethyl methacrylate (95). The group cultured both individual follicles and ovarian cortex tissues isolated from cats and dogs. The study evaluated the influence of flow on isolated ovarian follicles and effects of culture conditions on the follicular responsiveness to flow. This system enabled the study of mechanistic aspects of folliculogenesis, in particular primordial follicle activation and the interaction between microenvironment rigidity and follicle development. The understanding from large mammalian animal models is more relevant to human fertility preservation techniques and help in the development of enhanced ART.
Another approach that utilizes microfluidic technology for the purpose of fertility preservation has been described by He (2017), who used a nonplanar PDMS device to study microfluidic encapsulation of rodent ovarian follicles in core-shell hydrogel microcapsules (96). Their study showed that this technique could create biomimetic ovarian microtissue that recapitulated the mechanical heterogeneity in the extracellular matrix, which is essential to regulate follicle development. Thus, this technique could be used to facilitate follicle culture as a part of ART aiming to preserve the fertility of women.
Many current ARTs have operator-to-operator variation and lack standardization due to their reliance on skilled embryologists (97). For instance, ART clinics use enzymatic action of hyaluronidase to denude oocytes from the cumulus cell-corona cell mass that surround the oocyte (98). This involves the labor of mechanical pipetting and risks damaging the oocyte (99). A microfluidic device capable of denuding oocytes from the surrounding cumulus-corona cell mass in a continuous fluid flow in order to overcome these barriers was demonstrated by Weng et al (2018) (100). The group used standard PDMS soft lithography techniques to fabricate a microfluidic device that employed repeating constriction-expansion units of optimized geometries and surface features to completely denudate mouse oocytes. Additionally, their device did not use excessive mechanical stress, and hence it maintained developmental potential of the oocyte. Such devices have invaluable translational potential and could result in an automated and standardized processing of human samples in a clinical setting.
Microfluidic Modeling of the Fallopian Tube
The fallopian tubes in humans, called oviducts in other mammals, are tubes that originate near the ovaries and ultimately transition into the uterus. The epithelial lining of the fallopian tube consists of ciliated and secretory cells that make close contact with the gametes and early embryo (101-103). Additionally, the intricately folded tubular morphology, muscle contractions, and ciliary beating of these tubes largely influence fluid flux and movement of sperm and oocyte, enabling fertilization (104-107). In addition to providing a passageway for gametes and early-stage embryos to travel, the fallopian tubes also provide an optimal microenvironment for the embryo’s preimplantation development and epigenetic reprogramming (108-110).
Anatomical inaccessibility and difficulty in replicating in vivo gradients create challenges to studying the signaling pathways and processes of the fallopian tube that contribute to fertilization and developmental competence of the early embryo. Currently, in vitro oviductal or fallopian tubes models have used monolayer cultures of oviduct epithelial cells (111-113). However, fallopian tube cultures on plastic can undergo a rapid transformation of the differentiated, cuboidal–columnar oviduct epithelial cells into flattened cells, thus losing some of their cilia and secretory ability, compromising aspects of the in vivo tubular physiology (114, 115). Hence, a microfluidic model that maintains cell differentiation is beneficial in investigating the factors, such as hormones, growth factors, and shear stress that influence fertilization and preimplantation development.
Ferraz et al (2018) designed PDMS microfluidic device that maintained the in vivo–like morphology of bovine oviduct epithelial cells including a cuboidal to columnar pseudostratified epithelium with ciliated and secretory cells and function (114). In addition to supporting cell growth and differentiation similarly to that observed in vivo, their oviduct-on-a-chip model also produced bovine zygotes with a transcriptome and global methylation pattern similar to that of zygotes in vivo. This device provides a physiological microenvironment ideal for advancing our knowledge of gamete interaction, fertilization, embryogenesis, and zygote genetic reprogramming. Additionally, the device possesses a live imaging feature that can be useful in assessing the effects of drugs on gametes or embryos in real time.
In addition to its use in studying ovaries, the Solo-MFP designed by Xiao et al (2017) was also employed by Jackson-Bey et al (2020) to study the effects of high testosterone on function of human fallopian tube epithelium (116). Using the microfluidic device, the group cultured human fallopian tube epithelial tissue obtained from women undergoing salpingectomy. In addition to maintaining viability, the tissue retained cilia beating activity for 14 days. The study found that increased exposure to testosterone modifies expression of multiple genes that regulate cilia beating. These findings demonstrate the application of microfluidic systems in elucidating the mechanisms of subfertility in women living with hyperandrogenic disorders.
Microfluidic Modeling of the Uterus and Endometrium
The uterus is a muscular organ consisting of the corpus and cervix. The endometrium is 1 of 3 layers that lines the uterine cavity and its thickness varies throughout the menstrual cycle (2, 69). After ovulation, during the luteal phase, if the egg is fertilized by sperm, the resulting embryo implants into the endometrium. The uterus then provides mechanical protection and nutritional support to the embryo until delivery. However, if conception does not occur, the endometrial tissue of the uterine lining is discharged during menstruation. After menstruation, the endometrial tissue repairs, grows, and differentiates to prepare for the next cycle (69, 117).
Depending on the cyclic stimulation of ovarian sex steroids, endometrial stromal fibroblasts and vascular endothelial cells undergo morphological and biochemical changes to support embryo implantation and vascular function regulation (118). Various processes are involved in preparing the endometrial tissue for reproduction, but the details of these processes remain unclear (119). Decidualization of endometrial stoma is one such process that is critical for the successful establishment and maintenance of pregnancy to term (120, 121). This transformation involves differentiation of stromal fibroblasts in response to sex steroids and is marked by a transition to an epithelial-like cuboidal cell shape and secretion of pro-gestational proteins (122, 123). Despite the importance of decidualization in establishing embryo implantation, it has been difficult to investigate the triggering mechanism and the early stages of this process in a static culture due to the lack of vascular perfusion (124, 125). Hence, a dynamic microfluidic model of the endothelium may lead to a better understanding of the interaction between decidualization, pharmaceutical agents, or environmental toxicants, and fertility.
Gnecco et al (2019) designed a model of the endometrial perivascular stroma by integrating a porous membrane in a dual chamber microfluidic device in PDMS (126). The porous membrane enabled diffusion of small molecules as well as high-resolution bright field and fluorescent imaging. This model sustained an in vitro co-culture of human endometrial stromal and endothelial cells for 4 weeks. While stimulating the hormonal changes associated with the human menstrual cycle, the group observed successful differentiation of stromal cells into functional decidual cells. Additionally, the group controlled the microfluidic properties of the device to subject the cells to experimental laminar perfusion that mimics the hemodynamic forces derived from endometrial blood flow. They found that hemodynamic forces induced secretion of specific endothelial cell-derived prostanoids that enhanced endometrial perivascular decidualization. This device has further potential to identify bidirectional crosstalk between cells and hemodynamics that may cause endometriosis, pregnancy failure, and infertility.
In addition to the uterine vascularization, the quality of the embryo is also influenced by postimplantation processes (127). The initial postimplantation morphological development in human embryo is marked by the apical–basal polarization and lumenogenesis of the epiblast that results in the pro-amniotic cavity (128, 129). However, there are barriers to studying these processes. Maintaining human embryos in vitro beyond the blastocyst stage remains challenging due to suboptimal protocols (130, 131). Moreover, bioethical guidelines restrict in vitro culture of human embryos to either 14 days postfertilization or to the onset of primitive streak development (132). Microfluidic systems can help in study of postimplantation developmental processes as shown by a human pluripotent stem cells (hPSCs)-based device developed by Zheng at al (2019) (133). Geltrex was preloaded in the device and the material contracted during gelation, forming concave gel pockets that acted as a 3D environment for cells loaded in the device. The model successfully recapitulated early postimplantation developmental events, such as lumenogenesis of the epiblast, differentiation of primordial germ cells, and formation of pro-amniotic cavity and embryonic sac. The control and scalability offered by their microfluidic model makes it an apt tool for investigating the influence of drugs and toxins on pregnancy.
When studying drug safety and efficacy, it is important to recognize that the absorption, distribution, metabolism, excretion, and toxicity of a drug is dictated by multiple cell types that are linked by intricate mechanisms (134). Edington et al 2018 fabricated a microfluidic platform with 10 organs: liver, lung, gut, endometrium, brain, heart, pancreas, kidney, skin, and skeletal muscle (135). The platform comprises 3 layers that involve a pneumatic bottom layer, a fluidic top layer, and an intermediate membrane. The top layer was made from a polysulfone plastic and the intermediate membrane was made of polyurethane. The platform sustained the phenotypic functionality of all 10 modules for 4 weeks and the circulating media flow allowed them to interact and exchange endogenously produced molecules. Because one of the organs included was the endometrium, the platform can be employed for studying pharmacokinetics during drug discovery as well as for disease modeling.
Microfluidic Modeling of Placenta
The placenta is a highly specialized organ in the human body and is composed of trophoblasts, connective tissue, basal lamina, and the fetal endothelium (136). The placenta is critical to a successful pregnancy as it acts as a barrier between the fetal circulation and maternal blood, and thus, it tightly regulates the exchange of endogenous and exogenous substances between the mother and the fetus. The lack of understanding about this key organ’s inner workings is a barrier to elucidating the effects of maternally administered drugs on the developing fetus. Additionally, a range of complications of pregnancy, such as preeclampsia and intrauterine growth restriction, have been linked to abnormalities in placenta structure and function (137, 138).
Traditionally, in vitro and ex vivo models that mimic physiological drug transport across the maternal-fetal interface in the human placenta have been inadequate (139). In vitro models used have been based on static culture of trophoblast monolayers in Transwell inserts and ex vivo models used extracorporeal perfusion of cannulated whole human placenta (140-143). While some toxicology studies have used animal models of pregnancy, there are considerable interspecies differences in placenta, and the 1960s tragedy of thalidomide raised questions about the predictive value of animal data (144). To better assess the safety of medications in pregnancy, one option would be to screen drug compounds for their ability to cross the placenta using a microfluidic model that captures the multilayered architecture, intercellular junctions, and hemodynamic environment of the placental barrier.
Blundell et al (2018) created a placenta-on-a-chip model by setting up a co-culture of human trophoblasts and human placental villous endothelial cells that grew in apposition on a fibronectin-coated semipermeable membrane in a compartmentalized microfluidic device (145). By precisely manipulating various key control parameters, the group was able to account for biochemical and physical factors that influence the dynamics of placental transfer such as utero-placental and umbilical blood flow, barrier thickness, and concentration gradients (146). The continuous perfusion and dynamic culture conditions of their model maintained cell viability and reproduced native morphological characteristics of the placenta such as generation of microvilli. To portray the applicability of their device to investigate placental drug transport, the group provided proof-of-principle for the role of breast cancer resistance protein (BCRP; an efflux transporter highly expressed on trophoblast cells) in mediating transport of glyburide, an oral drug used to treat diabetes in pregnancy (147). As the device is able to replicate pathological features of different placental disorders, it has the potential to serve as an in vitro disease model that can be used to develop new therapeutic strategies.
In addition to drugs, a microfluidic placenta model can also be used to analyze the interaction and transport of environmental contaminants and food additives through the placenta. A good example is environmental nanomaterials, which are prevalent due to the emerging nanotechnology industry (148, 149). Pregnant women are likely to encounter these particles through air or water. Yin et al (2019) investigated placental responses to environmental nanomaterials by co-culturing a human choriocarcinoma cell line and human umbilical vein endothelial cells on a PDMS chip (150). Additionally, through the addition of extracellular matrix using Matrigel, the group replicated in vivo microenvironment on the chip. Once the morphology and functionality of the model was confirmed, they exposed the model to titanium dioxide nanoparticles and found increased permeability due to ruptures in the placental barrier and impaired maternal immune cell behavior.
The placenta barrier facilitates gas exchange to the fetus, ensuring that the fetus is supplied adequate levels of oxygen and that carbon dioxide is removed (151). Hence, microfluidic models of placenta have clinical applicability in neonatal respiratory care of infants born prematurely that often have structurally and functionally immature lungs. Currently, these infants are given mechanical ventilation or extracorporeal membrane oxygenation. However, these can cause long-term complications or even death (152, 153). The premature transition from liquid to gas ventilation puts these infants at risk of breathing disorders such as bronchopulmonary dysplasia (154). Hence, an approach where extracorporeal gas exchange is maintained by the placenta is preferred.
Partridge et al (2017) created an extracorporeal device that was able to support an extremely premature fetal lamb for 4 weeks without apparent physiologic derangement or organ failure (155). They overcame the obstacle of circulatory failure due to preload or afterload imbalance imposed on the fetal heart by oxygenator resistance and pump-supported circuits by using a pumpless arteriovenous circuit along with a closed fluid environment that continuously exchanged fluid. This fluid exchange ensured nutritional and oxygen supply to the fetus, mimicking amniotic fluid. As such, this device may eventually not only be used to support premature infants but also to directly administer therapeutic agents to the isolated fetus.
Microfluidic Modeling of Diseases and Toxic Exposures
In addition to understanding reproductive processes and enhancing ART, microfluidic models of the female reproductive tract also have an application in studying exposures to EDCs as well as diseases of the female reproductive tract such as ovarian cancer and preeclampsia.
Ovarian Cancer
With a 10-year survival rate of <30%, ovarian cancer is one of the 5 leading causes of cancer death in females, likely due to insufficient early detection methods (156-158). The most common histotype of ovarian cancer is high-grade serous carcinomas (HGSC) and it is derived from the epithelial cells of the fallopian tube (156, 159, 160). One potential diagnostic strategy that incorporates the use of microfluidics is to utilize extracellular vesicles derived from biofluids for early and noninvasive detection of the disease.
A microfluidic-based device from Dorayappan et al (2019) isolated exosomes from culture media and cancer patient samples in a cost-effective and highly specific manner (161). Mass spectroscopy of these samples allowed them to identity exosome cargo proteins that are differentially expressed in HGSC cell lines compared with normal cells. These protein biomarkers may serve as part of an early detection strategy for cancer using biofluids. In fact, Zhang et al (2019) demonstrated using their microfluidic system termed MINDS (multiscale integration by designed self-assembly) that elevated folate receptor alpha (FRα) protein in ovarian cancer extracellular vesicles can be used as a biomarker for liquid biopsy-based cancer diagnosis (162). Their system integrated micro-patterning and 3D nanostructured herringbone mixer for molecular recognition and flow manipulation to detect ovarian cancer using as little as 2 microliters of plasma.
Ferraz et al (2020) created an oviduct-on-a-chip model using dog oviduct tissue discarded during routine ovary-hysterectomy (163). The model was responsive to hormonal stimulation patterns associated with the menstrual cycle. Moreover, using CRISPR-Cas9 to knock out the oncogene p53, the group was able to recapitulate serous tubal intraepithelial carcinoma, a premetastatic form of HGSC, as well as events associated with human serous tubal intraepithelial carcinoma, such as decreased ciliation, loss of cell polarization, and increased cell proliferation. In vitro models like these could help study early-stage events and identify novel biomarkers of ovarian cancer.
Preeclampsia
Preeclampsia is a hypertensive pregnancy complication that affects approximately 3.4% of pregnancies in the United States (164, 165). The disease is characterized by high maternal blood pressure that decreases blood supply, reducing fetal access to oxygen and nutrients. This condition increases the risk for organ failure, stroke, preterm birth, and stillbirth. Women who opt for ART such as egg donation, donor insemination, or in vitro fertilization are at a higher risk for preeclampsia (166-168). The disease has been associated with higher levels of circulating and placental soluble fms-like tyrosine kinase-1 (sFlt-1), an anti-angiogenic protein that disrupts angiogenic balance by antagonizing the proangiogenic factor, vascular endothelial growth factor (VEGF) (169). Use of microfluidic systems in preeclampsia research can open new avenues for treatment as shown by Trapiella-Alfonso et al (2019) (170). The group suggested restoring the angiogenic balance in patients by using VEGF-coated magnetic beads to capture sFlt-1 and release phosphatidylinositol-glycan biosynthesis class F protein (PIGF), a proangiogenic factor. The group utilized a microfluidic device to mimic a small-scale extracorporeal circulation system to assess the validity of their concept and they found that their approach resulted in a 40% reduction of sFlt-1 and up to 2-fold increase of free PIGF. Thus, microfluidic systems can be used to validate proof-of-concepts of new therapeutic approaches for preeclampsia.
Endocrine Disrupting Chemicals
EDCs are a class of chemicals that can mimic, block, or interfere with hormones in the body’s endocrine system due to similarities in their chemical structures (171). These are present in the environment as plastics (bisphenols), plasticizers (phthalates), fungicides (vinclozolin), and pharmaceutical agents (diethylstilbestrol) (172). Recently, it has been found that these chemicals, even in small amounts, can exert toxic effects on the female reproductive system and adversely affect a woman’s reproductive outcomes (173). Specifically, bisphenols have been suggested to hinder with uterine development and reproductive function by interacting with estrogen receptors and altering expression of estrogen-regulated genes (174-178). EDCs have also been reported in higher levels in patients with polycystic ovarian syndrome (179, 180).
The tunability of microfluidic systems offers the ability to characterize the effects of EDC exposure. The devices described in this review can be manipulated to examine the effects of EDC on specific organs such as ovaries, fallopian tubes, endometrium, and placenta as well as on physiological processes of ovulation, fertilization, implantation, embryogenesis, and placental transport. Given the complexity of effects resulting from EDC exposure, coupled with the high level of interaction between the organs of the reproductive system, a microfluidic device that cultures an interconnected network of multiple organs would be more informative about the collective response to EDCs.
The device termed “EVATAR” is one such platform that is capable of supporting 5 interconnected organs and uses embedded electromagnetic actuation (77). Xiao et al (2017) employed this device with co-cultured human liver spheroids, mouse ovarian explants, human fallopian tube epithelium, human endometrium, and human cervix tissues. The group was successfully able to maintain tissue viability and hormonal responsiveness for 28 days. By introducing liver on the platform, the potential toxicity of EDC metabolism products can also be investigated.
Future Directions and Challenges
While microfluidic systems have made many advancements that were previously challenging with static cultures, there are still hurdles to overcome. For instance, further development is needed to improve the material used, fabrication processes, and other practical aspects of the technology.
The fabrication of microfluidic devices often requires access to specialized cleanroom facilities. The EVATAR system assembly needs special equipment, as the process involves producing an intermediate thermoplastic track-etched membrane from materials such as polycarbonate or polyethylene terephthalate (47). An alternative popular material is PDMS which has a user-friendly protocol, optical clarity, and requires commonly available lab equipment (181). However, PDMS has the tendency to absorb hydrophobic molecules, specifically hormones, making its applicability in reproductive research questionable (182, 183). The lack of alternatives and the ability to perform shorter experiments with certain coatings of the PDMS has made it acceptable for some studies (184-186). However, when studying the altered response of biological processes due to exposure to drugs, xenobiotics, pathogens, and environmental toxicants, it is important to quantify and account for potential material–compound absorption or interaction before concluding the findings.
Another popular material for constructing microfluidic devices is polysulfone (PSF) (135). This rigid machinable thermoplastic offers several advantages over PDMS and is often used in medical devices since it has US Food and Drug Administration (FDA) food grade approval (21CFR177.1655) as well as United States Pharmacopeia (USP) Class VI biocompatibility (187). The material is easy to sterilize as it can be autoclaved and is impenetrable to chemical solvents (188). Compared with most elastomers such as PDMS, polysulfone has significantly lower adsorption and absorption of hydrophobic compounds, making it suitable for studies involving drugs and hormones (189). While the material lacks the optical clarity and oxygen permeability of PDMS, neither of these factors are significant challenges as most microfluidic systems are open and have an air-liquid interface where gas exchange can occur.
Each microfluidic device produces a different fluid flow and shear stress profile. There is a large amount of optimization required to determine the ideal settings and configurations for replicating an organ’s in vivo structure and microenvironment. Moreover, the technical details become more challenging when culturing multiple cell lines or tissues on a single platform, as this creates a need to find a flow rate, sample volume, and scaling for the size of tissues. Research is needed into developing a media composition that is compatible with multiple organ model systems or cell types, as media needs to be circulated throughout the device to ensure interaction through secreted factors and hormones.
Conclusion
Studying the physiology of a system as intricate as the female reproductive tract is challenging. Microfluidic systems integrate engineering and biology to revolutionize cell culturing and experimenting techniques. The ability to integrate cellular and tissue level cytokine and endocrine loops into signaling pathways and to engineer precise temporal manipulations of microliter volume of fluids and hormones allows the reproducible study of the female reproductive tract in a physiologically relevant setting. While progress is required to optimize the use of microfluidic systems, this technology presents a unique opportunity to study human embryology, reproduction, and disorders of the female reproductive tract.
Acknowledgments
The authors wish to acknowledge financial support through grants UH3 ES029073 and CA 240301 from the National Institutes of Health (NIH), Bethesda, Maryland. V.V.B. is a recipient of the University of Illinois at Chicago Graduate College University Fellowship. Figures were created with BioRender.com.
Glossary
Abbreviations
- 3D
3-dimensional
- ART
assisted reproductive technologies
- EDC
endocrine disrupting chemical
- HGSC
high-grade serous carcinoma
- PDMS
polydimethylsiloxane
- PIGF
phosphatidylinositol-glycan biosynthesis class F protein
- sFlt-1
soluble fms-like tyrosine kinase-1
Additional Information
Disclosures: The authors have no conflicts of interest to disclose.
Data Availability
Data sharing is not applicable to this article because no data sets were generated or analyzed during the present study.
References
- 1. Kurita T, Wang YZ, Donjacour AA, et al. Paracrine regulation of apoptosis by steroid hormones in the male and female reproductive system. Cell Death Differ. 2001;8(2):192-200. [DOI] [PubMed] [Google Scholar]
- 2. Reis FM, Cobellis L, Luisi S, et al. Paracrine/autocrine control of female reproduction. Gynecol Endocrinol. 2000;14(6):464-475. [DOI] [PubMed] [Google Scholar]
- 3. Brinkley HJ. Endocrine Signaling and Female Reproduction. Biol Reprod. 1981;24(1):22-43. [DOI] [PubMed] [Google Scholar]
- 4. Malcomson RDG, Nagy A. The endocrine system. In: Khong T.Y., Malcomson R.D.G., eds. Keeling’s Fetal Neonatal Pathol. Springer, Cham; 2015:671-702. 10.1007/978-3-319-19207-9_25 [DOI] [Google Scholar]
- 5. Nguyen PV, Kafka JK, Ferreira VH, Roth K, Kaushic C. Innate and adaptive immune responses in male and female reproductive tracts in homeostasis and following HIV infection. Cell Mol Immunol. 2014;11(5):410-427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Buyuk E, Nejat E, Neal-Perry G. Determinants of female reproductive senescence: differential roles for the ovary and the neuroendocrine axis. Semin Reprod Med. 2010;28(5):370-379. [DOI] [PubMed] [Google Scholar]
- 7. dos Santos RL, da Silva FB, Ribeiro RF Jr, Stefanon I. Sex hormones in the cardiovascular system. Horm Mol Biol Clin Investig. 2014;18(2):89-103. [DOI] [PubMed] [Google Scholar]
- 8. Clarke BL, Khosla S. Female reproductive system and bone. Arch Biochem Biophys. 2010;503(1):118-128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Jin M, Yu Y, Huang H. An update on primary ovarian insufficiency. Sci China Life Sci. 2012;55(8):677-686. [DOI] [PubMed] [Google Scholar]
- 10. Goswami D, Conway GS. Premature ovarian failure. Hum Reprod Update. 2005;11(4):391-410. [DOI] [PubMed] [Google Scholar]
- 11. Tao JJ, Visvanathan K, Wolff AC. Long term side effects of adjuvant chemotherapy in patients with early breast cancer. Breast. 2015;24(Suppl 2):S149-S153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Aziz AUR, Yu X, Jiang Q, et al. Doxorubicin-induced toxicity to 3D-cultured rat ovarian follicles on a microfluidic chip. Toxicol In Vitro. 2020;62:104677. [DOI] [PubMed] [Google Scholar]
- 13.Centers for Disease Control and Prevention. Common Reproductive Health Concerns for Women. Reviewed April 27, 2018.Accessed December 27, 2020. https://www.cdc.gov/reproductivehealth/womensrh/healthconcerns.html
- 14.National Center for Health Statistics. NSFG - Listing I - Key Statistics from the National Survey of Family Growth. Reviewed November 8, 2019. Accessed December 27, 2020.https://www.cdc.gov/nchs/nsfg/key_statistics/i_2015-2017.htm#infertility
- 15. Adamson GD, Tabangin M, Macaluso M, de Mouzon J. The number of babies born globally after treatment with the assisted reproductive technologies (ART). Fertil Steril. 2013;100(3):S42. [Google Scholar]
- 16. Pinborg A. Short- and long-term outcomes in children born after assisted reproductive technology. Bjog. 2019;126(2):145-148. [DOI] [PubMed] [Google Scholar]
- 17. Asghar W, El Assal R, Shafiee H, Anchan RM, Demirci U. Preserving human cells for regenerative, reproductive, and transfusion medicine. Biotechnol J. 2014;9(7):895-903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Smitz I, Dolmans MM, Donnez J, et al. Current achievements and future research directions in ovarian tissue culture, in vitro follicle development and transplantation: implications for fertility preservation. Hum Reprod Update. 2010;16(4):395-414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Servick K. Unsettled questions trail IVF’s success. Science. 2014;345(6198):744-746. [DOI] [PubMed] [Google Scholar]
- 20. Li T, Vu TH, Ulaner GA, et al. IVF results in de novo DNA methylation and histone methylation at an Igf2-H19 imprinting epigenetic switch. Mol Hum Reprod. 2005;11(9):631-640. [DOI] [PubMed] [Google Scholar]
- 21. Huntriss J, Picton HM. Epigenetic consequences of assisted reproduction and infertility on the human preimplantation embryo. Hum Fertil (Camb). 2008;11(2):85-94. [DOI] [PubMed] [Google Scholar]
- 22. Havelock JC, Rainey WE, Carr BR. Ovarian granulosa cell lines. Mol Cell Endocrinol. 2004;228(1-2):67-78. [DOI] [PubMed] [Google Scholar]
- 23. Ince TA, Sousa AD, Jones MA, et al. Characterization of twenty-five ovarian tumour cell lines that phenocopy primary tumours. Nat Commun. 2015;6:7419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12(4):207-218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Murphy AR, Wiwatpanit T, Lu Z, Davaadelger B, Kim JJ. Generation of Multicellular Human Primary Endometrial Organoids. J Vis Exp. 2019;(152):e60384. [DOI] [PMC free article] [PubMed]
- 26. Duval K, Grover H, Han LH, et al. Modeling Physiological Events in 2D vs. 3D Cell Culture. Physiology (Bethesda). 2017;32(4):266-277. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 27. Xu M, West-Farrell ER, Stouffer RL, Shea LD, Woodruff TK, Zelinski MB. Encapsulated three-dimensional culture supports development of nonhuman primate secondary follicles. Biol Reprod. 2009;81(3):587-594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Eddie SL, Quartuccio SM, Zhu J, et al. Three-dimensional modeling of the human fallopian tube fimbriae. Gynecol Oncol. 2015;136(2):348-354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Tibbitt MW, Anseth KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol Bioeng. 2009;103(4):655-663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Pampaloni F, Reynaud EG, Stelzer EHK. The third dimension bridges the gap between cell culture and live tissue. Nat Rev Mol Cell Biol. 2007;8(10):839-845. [DOI] [PubMed] [Google Scholar]
- 31. Su YQ, Sugiura K, Eppig JJ. Mouse oocyte control of granulosa cell development and function: paracrine regulation of cumulus cell metabolism. Semin Reprod Med. 2009;27(1):32-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Eppig JJ, Pendola FL, Wigglesworth K, Pendola JK. Mouse oocytes regulate metabolic cooperativity between granulosa cells and oocytes: amino acid transport. Biol Reprod. 2005;73(2):351-357. [DOI] [PubMed] [Google Scholar]
- 33. Kreeger PK, Fernandes NN, Woodruff TK, Shea LD. Regulation of mouse follicle development by follicle-stimulating hormone in a three-dimensional in vitro culture system is dependent on follicle stage and dose. Biol Reprod. 2005;73(5):942-950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Su YQ, Sugiura K, Wigglesworth K, et al. Oocyte regulation of metabolic cooperativity between mouse cumulus cells and oocytes: BMP15 and GDF9 control cholesterol biosynthesis in cumulus cells. Development. 2008;135(1):111-121. [DOI] [PubMed] [Google Scholar]
- 35. Gilchrist RB, Ritter LJ, Myllymaa S, et al. Molecular basis of oocyte-paracrine signalling that promotes granulosa cell proliferation. J Cell Sci. 2006;119(Pt 18):3811-3821. [DOI] [PubMed] [Google Scholar]
- 36. Baker BM, Trappmann B, Stapleton SC, Toro E, Chen CS. Microfluidics embedded within extracellular matrix to define vascular architectures and pattern diffusive gradients. Lab Chip. 2013;13(16):3246-3252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Lelièvre SA, Kwok T, Chittiboyina S. Architecture in 3D cell culture: an essential feature for in vitro toxicology. Toxicol In Vitro. 2017;45(3):287-295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Sung JH, Esch MB, Prot JM, et al. Microfabricated mammalian organ systems and their integration into models of whole animals and humans. Lab Chip. 2013;13(7):1201-1212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Moya ML, Hsu YH, Lee AP, Hughes CC, George SC. In vitro perfused human capillary networks. Tissue Eng Part C Methods. 2013;19(9):730-737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Chen MB, Whisler JA, Jeon JS, Kamm RD. Mechanisms of tumor cell extravasation in an in vitro microvascular network platform. Integr Biol (Camb). 2013;5(10):1262-1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Bischel LL, Young EW, Mader BR, Beebe DJ. Tubeless microfluidic angiogenesis assay with three-dimensional endothelial-lined microvessels. Biomaterials. 2013;34(5):1471-1477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Wikswo JP, Curtis EL, Eagleton ZE, et al. Scaling and systems biology for integrating multiple organs-on-a-chip. Lab Chip. 2013;13(18):3496-3511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Sosa-Hernández JE, Villalba-Rodríguez AM, Romero-Castillo KD, et al. Organs-on-a-chip module: a review from the development and applications perspective. Micromachines. 2018;9(10):536. doi: 10.3390/mi9100536 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Ingber DE. Mechanobiology and diseases of mechanotransduction. Ann Med. 2003;35(8):564-577. [DOI] [PubMed] [Google Scholar]
- 45. Mammoto T, Mammoto A, Ingber DE. Mechanobiology and developmental control. Annu Rev Cell Dev Biol. 2013;29:27-61. [DOI] [PubMed] [Google Scholar]
- 46. Takayama S, Ostuni E, LeDuc P, Naruse K, Ingber DE, Whitesides GM. Subcellular positioning of small molecules. Nature. 2001;411(6841):1016. [DOI] [PubMed] [Google Scholar]
- 47. Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol. 2014;32(8):760-772. [DOI] [PubMed] [Google Scholar]
- 48. Ahadian S, Civitarese R, Bannerman D, et al. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. [published correction appears in Adv Healthc Mater. 2018 Jul;7(14):]. Adv Healthc Mater. 2018;7(2): 10.1002/adhm.201700506. doi:10.1002/adhm.201700506 [DOI] [PubMed] [Google Scholar]
- 49. Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science. 2010;328(5986):1662-1668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Heylman C, Sobrino A, Shirure VS, Hughes CC, George SC. A strategy for integrating essential three-dimensional microphysiological systems of human organs for realistic anticancer drug screening. Exp Biol Med (Maywood). 2014;239(9):1240-1254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Sung JH, Shuler ML. A micro cell culture analog (CCA) with 3-D hydrogel culture of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer drugs. Lab Chip. 2009;9(10):1385-1394. [DOI] [PubMed] [Google Scholar]
- 52. Ronaldson-Bouchard K, Vunjak-Novakovic G. Organs-on-a-Chip: A Fast Track for Engineered Human Tissues in Drug Development. Cell Stem Cell. 2018;22(3):310-324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Nawroth JC, Lucchesi C, Cheng D, et al. A Microengineered Airway Lung Chip Models Key Features of Viral-induced Exacerbation of Asthma. Am J Respir Cell Mol Biol. 2020;63(5):591-600. [DOI] [PubMed] [Google Scholar]
- 54. Grassart A, Malardé V, Gobaa S, et al. Bioengineered Human Organ-on-Chip Reveals Intestinal Microenvironment and Mechanical Forces Impacting Shigella Infection. Cell Host Microbe. 2019;26(4):565. [DOI] [PubMed] [Google Scholar]
- 55. Benam KH, Novak R, Nawroth J, et al. Matched-Comparative Modeling of Normal and Diseased Human Airway Responses Using a Microengineered Breathing Lung Chip. Cell Syst. 2016;3(5):456-466.e4. [DOI] [PubMed] [Google Scholar]
- 56. Kasendra M, Luc R, Yin J, et al. Duodenum intestine-chip for preclinical drug assessment in a human relevant model. Elife. 2020;9:1-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Jang KJ, Otieno MA, Ronxhi J, et al. Reproducing human and cross-species drug toxicities using a Liver-Chip. Sci Transl Med. 2019;11(517):eaax5516. [DOI] [PubMed] [Google Scholar]
- 58. Jain A, Barrile R, van der Meer AD, et al. Primary Human Lung Alveolus-on-a-chip Model of Intravascular Thrombosis for Assessment of Therapeutics. Clin Pharmacol Ther. 2018;103(2):332-340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Kasendra M, Tovaglieri A, Sontheimer-Phelps A, et al. Development of a primary human Small Intestine-on-a-Chip using biopsy-derived organoids. Sci Rep. 2018;8(1):2871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Weber EJ, Chapron A, Chapron BD, et al. Development of a microphysiological model of human kidney proximal tubule function. Kidney Int. 2016;90(3):627-637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Van Ness KP, Chang SY, Weber EJ, Zumpano D, Eaton DL, Kelly EJ. Microphysiological Systems to Assess Nonclinical Toxicity. Curr Protoc Toxicol. 2017;73:14.18.1-14.18.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Nieskens TTG, Persson M, Kelly EJ, Sjögren AK. A Multicompartment Human Kidney Proximal Tubule-on-a-Chip Replicates Cell Polarization-Dependent Cisplatin Toxicity. Drug Metab Dispos. 2020;48(12):1303-1311. [DOI] [PubMed] [Google Scholar]
- 63. Pedrosa ML, Furtado MH, Ferreira MCF, Carneiro MM. Sperm selection in IVF: the long and winding road from bench to bedside. JBRA Assist Reprod. 2020;24(3):332-339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Marzano G, Chiriacò MS, Primiceri E, et al. Sperm selection in assisted reproduction: a review of established methods and cutting-edge possibilities. Biotechnol Adv. 2020;40:107498. [DOI] [PubMed] [Google Scholar]
- 65. Suarez SS, Wu M. Microfluidic devices for the study of sperm migration. Mol Hum Reprod. 2017;23(4):227-234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Samuel R, Feng H, Jafek A, Despain D, Jenkins T, Gale B. Microfluidic-based sperm sorting & analysis for treatment of male infertility. Transl Androl Urol. 2018;7(Suppl 3):S336-S347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Nosrati R, Graham PJ, Zhang B, et al. Microfluidics for sperm analysis and selection. Nat Rev Urol. 2017;14(12):707-730. [DOI] [PubMed] [Google Scholar]
- 68. Vaughan DA, Sakkas D. Sperm selection methods in the 21st century. Biol Reprod. 2019;101(6):1076-1082. [DOI] [PubMed] [Google Scholar]
- 69. Hawkins SM, Matzuk MM. The menstrual cycle: basic biology. Ann N Y Acad Sci. 2008;1135:10-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Woodruff TK. Making eggs: is it now or later? Nat Med. 2008;14(11):1190-1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Jeruss JS, Woodruff TK. Preservation of fertility in patients with cancer. N Engl J Med. 2009;360(9):902-911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Waimey KE, Smith BM, Confino R, Jeruss JS, Pavone ME. Understanding Fertility in Young Female Cancer Patients. J Womens Health (Larchmt). 2015;24(10):812-818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Blumenfeld Z. Chemotherapy and fertility. Best Pract Res Clin Obstet Gynaecol. 2012;26(3):379-390. [DOI] [PubMed] [Google Scholar]
- 74. Imai A, Furui T. Chemotherapy-induced female infertility and protective action of gonadotropin-releasing hormone analogues. J Obstet Gynaecol. 2007;27(1):20-24. [DOI] [PubMed] [Google Scholar]
- 75. Fleischer RT, Vollenhoven BJ, Weston GC. The effects of chemotherapy and radiotherapy on fertility in premenopausal women. Obstet Gynecol Surv. 2011;66(4):248-254. [DOI] [PubMed] [Google Scholar]
- 76. Poorvu PD, Frazier AL, Feraco AM, et al. Cancer Treatment-Related Infertility: A Critical Review of the Evidence. JNCI Cancer Spectr. 2019;3(1):pkz008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Xiao S, Coppeta JR, Rogers HB, et al. A microfluidic culture model of the human reproductive tract and 28-day menstrual cycle. Nat Commun. 2017;8:14584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. West ER, Shea LD, Woodruff TK. Engineering the follicle microenvironment. Semin Reprod Med. 2007;25(4):287-299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Desai N, Alex A, AbdelHafez F, et al. Three-dimensional in vitro follicle growth: overview of culture models, biomaterials, design parameters and future directions. Reprod Biol Endocrinol. 2010;8:119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Picton HM, Harris SE, Muruvi W, Chambers EL. The in vitro growth and maturation of follicles. Reproduction. 2008;136(6):703-715. [DOI] [PubMed] [Google Scholar]
- 81. Jewgenow K, Göritz F. The recovery of preantral follicles from ovaries of domestic cats and their characterisation before and after culture. Anim Reprod Sci. 1995;39(4):285-297. [Google Scholar]
- 82. Jewgenow K. Role of media, protein and energy supplements on maintenance of morphology and DNA-synthesis of small preantral domestic cat follicles during short-term culture. Theriogenology. 1998;49(8):1567-1577. [DOI] [PubMed] [Google Scholar]
- 83. Hovatta O, Wright C, Krausz T, Hardy K, Winston RM. Human primordial, primary and secondary ovarian follicles in long-term culture: effect of partial isolation. Hum Reprod. 1999;14(10):2519-2524. [DOI] [PubMed] [Google Scholar]
- 84. Griffith OW, Chavan AR, Protopapas S, Maziarz J, Romero R, Wagner GP. Embryo implantation evolved from an ancestral inflammatory attachment reaction. Proc Natl Acad Sci U S A. 2017;114(32):E6566-E6575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Chen Y, Yang W, Shi X, Zhang C, Song G, Huang D. The Factors and Pathways Regulating the Activation of Mammalian Primordial Follicles in vivo. Front Cell Dev Biol. 2020;8:575706. doi:10.3389/fcell.2020.575706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Williams CJ. Signalling mechanisms of mammalian oocyte activation. Hum Reprod Update. 2002;8(4):313-321. [DOI] [PubMed] [Google Scholar]
- 87. Raheem KA. Cytokines, growth factors and macromolecules as mediators of implantation in mammalian species. Int J Vet Sci Med. 2018;6(Suppl):S6-S14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Russell DL, Robker RL. Molecular mechanisms of ovulation: co-ordination through the cumulus complex. Hum Reprod Update. 2007;13(3):289-312. [DOI] [PubMed] [Google Scholar]
- 89. Georgadaki K, Khoury N, Spandidos DA, Zoumpourlis V. The molecular basis of fertilization (Review). Int J Mol Med. 2016;38(4):979-986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Grive KJ, Freiman RN. The developmental origins of the mammalian ovarian reserve. Development. 2015;142(15):2554-2563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Skory RM, Xu Y, Shea LD, Woodruff TK. Engineering the ovarian cycle using in vitro follicle culture. Hum Reprod. 2015;30(6):1386-1395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Smitz J, Cortvrindt R. Oocyte in-vitro maturation and follicle culture: current clinical achievements and future directions. Hum Reprod. 1999;14(Suppl. 1):145-161. [DOI] [PubMed] [Google Scholar]
- 93. Reynaud K, de Lesegno CV, Chebrout M, Thoumire S, Chastant-Maillard S. Follicle population, cumulus mucification, and oocyte chromatin configuration during the periovulatory period in the female dog. Theriogenology. 2009;72(8):1120-1131. [DOI] [PubMed] [Google Scholar]
- 94. Reynaud K, Gicquel C, Thoumire S, et al. Folliculogenesis and morphometry of oocyte and follicle growth in the feline ovary. Reprod Domest Anim. 2009;44(2):174-179. [DOI] [PubMed] [Google Scholar]
- 95. Nagashima JB, El Assal R, Songsasen N, Demirci U. Evaluation of an ovary-on-a-chip in large mammalian models: species specificity and influence of follicle isolation status. J Tissue Eng Regen Med. 2018;12(4):e1926-e1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. He X. Microfluidic Encapsulation of Ovarian Follicles for 3D Culture. Ann Biomed Eng. 2017;45(7):1676-1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kashaninejad N, Shiddiky MJA, Nguyen NT. Advances in Microfluidics-Based Assisted Reproductive Technology: From Sperm Sorter to Reproductive System-on-a-Chip. Adv Biosyst. 2018;2(3):1700197. doi:10.1002/adbi.201700197 [Google Scholar]
- 98. Rienzi L, Balaban B, Ebner T, Mandelbaum J. The oocyte. Hum Reprod. 2012;27(Suppl. 1):2-21. [DOI] [PubMed] [Google Scholar]
- 99. Zeringue HC, Rutledge JJ, Beebe DJ. Early mammalian embryo development depends on cumulus removal technique. Lab Chip. 2005;5(1):86-90. [DOI] [PubMed] [Google Scholar]
- 100. Weng L, Lee GY, Liu J, Kapur R, Toth TL, Toner M. On-chip oocyte denudation from cumulus-oocyte complexes for assisted reproductive therapy. Lab Chip. 2018;18(24):3892-3902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Lyons RA, Saridogan E, Djahanbakhch O. The reproductive significance of human Fallopian tube cilia. Hum Reprod Update. 2006;12(4):363-372. [DOI] [PubMed] [Google Scholar]
- 102. Mahmood T, Saridogan E, Smutna S, Habib AM, Djahanbakhch O. The effect of ovarian steroids on epithelial ciliary beat frequency in the human Fallopian tube. Hum Reprod. 1998;13(11):2991-2994. [DOI] [PubMed] [Google Scholar]
- 103. Kölle S, Dubielzig S, Reese S, Wehrend A, König P, Kummer W. Ciliary transport, gamete interaction, and effects of the early embryo in the oviduct: ex vivo analyses using a new digital videomicroscopic system in the cow. Biol Reprod. 2009;81(2):267-274. [DOI] [PubMed] [Google Scholar]
- 104. Yaniz JL, Lopez-Gatius F, Hunter RH. Scanning electron microscopic study of the functional anatomy of the porcine oviductal mucosa. Anat Histol Embryol. 2006;35(1):28-34. [DOI] [PubMed] [Google Scholar]
- 105. Steinhauer N, Boos A, Günzel-Apel AR. Morphological changes and proliferative activity in the oviductal epithelium during hormonally defined stages of the oestrous cycle in the bitch. Reprod Domest Anim. 2004;39(2):110-119. [DOI] [PubMed] [Google Scholar]
- 106. Nelis H, D’Herde K, Goossens K, et al. Equine oviduct explant culture: a basic model to decipher embryo-maternal communication. Reprod Fertil Dev. 2014;26(7):954-966. [DOI] [PubMed] [Google Scholar]
- 107. Ezzati M, Djahanbakhch O, Arian S, Carr BR. Tubal transport of gametes and embryos: a review of physiology and pathophysiology. J Assist Reprod Genet. 2014;31(10):1337-1347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Ménézo Y, Guérin P, Elder K. The oviduct: a neglected organ due for re-assessment in IVF. Reprod Biomed Online. 2015;30(3):233-240. [DOI] [PubMed] [Google Scholar]
- 109. Buhi WC, Alvarez IM, Kouba AJ. Secreted proteins of the oviduct. Cells Tissues Organs. 2000;166(2):165-179. [DOI] [PubMed] [Google Scholar]
- 110. Ventura-Juncá P, Irarrázaval I, Rolle AJ, Gutiérrez JI, Moreno RD, Santos MJ. In vitro fertilization (IVF) in mammals: epigenetic and developmental alterations. Scientific and bioethical implications for IVF in humans. Biol Res. 2015;48(1):1-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Ouhibi N, Menezo Y, Benet G, Nicollet B. Culture of epithelial cells derived from the oviduct of different species. Hum Reprod. 1989;4(3):229-235. [DOI] [PubMed] [Google Scholar]
- 112. Aldarmahi A. Establishment and characterization of female reproductive tract epithelial cell culture. J Microsc Ultrastruct. 2017;5(2):105-110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Abe H, Hoshi H. Bovine oviductal epithelial cells: their cell culture and applications in studies for reproductive biology. Cytotechnology. 1997;23(1-33):171-183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Ferraz MAMM, Rho HS, Hemerich D, et al. An oviduct-on-a-chip provides an enhanced in vitro environment for zygote genome reprogramming. Nat Commun. 2018;9(1):4934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Sostaric E, Dieleman SJ, van de Lest CH, et al. Sperm binding properties and secretory activity of the bovine oviduct immediately before and after ovulation. Mol Reprod Dev. 2008;75(1):60-74. [DOI] [PubMed] [Google Scholar]
- 116. Jackson-Bey T, Colina J, Isenberg BC, et al. Exposure of human fallopian tube epithelium to elevated testosterone results in alteration of cilia gene expression and beating. Hum Reprod. 2020;35(9):2086-2096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Maybin JA, Critchley HO. Menstrual physiology: implications for endometrial pathology and beyond. Hum Reprod Update. 2015;21(6):748-761. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Gong X, Liu Y, Chen Z, Xu C, Lu Q, Jin Z. Insights into the paracrine effects of uterine natural killer cells. Mol Med Rep. 2014;10(6):2851-2860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Singh M, Chaudhry P, Asselin E. Bridging endometrial receptivity and implantation: network of hormones, cytokines, and growth factors. J Endocrinol. 2011;210(1):5-14. [DOI] [PubMed] [Google Scholar]
- 120. Garrido-Gomez T, Dominguez F, Quiñonero A, et al. Defective decidualization during and after severe preeclampsia reveals a possible maternal contribution to the etiology. Proc Natl Acad Sci U S A. 2017;114(40):E8468-E8477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Gellersen B, Brosens JJ. Cyclic decidualization of the human endometrium in reproductive health and failure. Endocr Rev. 2014;35(6):851-905. [DOI] [PubMed] [Google Scholar]
- 122. Gellersen B, Brosens IA, Brosens JJ. Decidualization of the human endometrium: mechanisms, functions, and clinical perspectives. Semin Reprod Med. 2007;25(6):445-453. [DOI] [PubMed] [Google Scholar]
- 123. Kennedy TG, Gillio-Meina C, Phang SH. Prostaglandins and the initiation of blastocyst implantation and decidualization. Reproduction. 2007;134(5):635-643. [DOI] [PubMed] [Google Scholar]
- 124. Gnecco JS, Pensabene V, Li DJ, et al. Compartmentalized Culture of Perivascular Stroma and Endothelial Cells in a Microfluidic Model of the Human Endometrium. Ann Biomed Eng. 2017;45(7):1758-1769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Arslan SY, Yu Y, Burdette JE, et al. Novel three dimensional human endocervix cultures respond to 28-day hormone treatment. Endocrinology. 2015;156(4):1602-1609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Gnecco JS, Ding T, Smith C, Lu J, Bruner-Tran KL, Osteen KG. Hemodynamic forces enhance decidualization via endothelial-derived prostaglandin E2 and prostacyclin in a microfluidic model of the human endometrium. Hum Reprod. 2019;34(4):702-714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Diedrich K, Fauser BC, Devroey P, Griesinger G; Evian Annual Reproduction (EVAR) Workshop Group . The role of the endometrium and embryo in human implantation. Hum Reprod Update. 2007;13(4):365-377. [DOI] [PubMed] [Google Scholar]
- 128. Taniguchi K, Shao Y, Townshend RF, et al. Lumen Formation Is an Intrinsic Property of Isolated Human Pluripotent Stem Cells. Stem Cell Reports. 2015;5(6):954-962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Shahbazi MN, Scialdone A, Skorupska N, et al. Pluripotent state transitions coordinate morphogenesis in mouse and human embryos. Nature. 2017;552(7684):239-243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Deglincerti A, Croft GF, Pietila LN, Zernicka-Goetz M, Siggia ED, Brivanlou AH. Self-organization of the in vitro attached human embryo. Nature. 2016;533(7602):251-254. [DOI] [PubMed] [Google Scholar]
- 131. Shahbazi MN, Jedrusik A, Vuoristo S, et al. Self-organization of the human embryo in the absence of maternal tissues. Nat Cell Biol. 2016;18(6):700-708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Daley GQ, Hyun I, Apperley JF, et al. Setting Global Standards for Stem Cell Research and Clinical Translation: The 2016 ISSCR Guidelines. Stem Cell Reports. 2016;6(6):787-797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Zheng Y, Xue X, Shao Y, et al. Controlled modelling of human epiblast and amnion development using stem cells. Nature. 2019;573(7774):421-425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Tsamandouras N, Chen WLK, Edington CD, Stokes CL, Griffith LG, Cirit M. Integrated Gut and Liver Microphysiological Systems for Quantitative In Vitro Pharmacokinetic Studies. Aaps J. 2017;19(5):1499-1512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Edington CD, Chen WLK, Geishecker E, et al. Interconnected Microphysiological Systems for Quantitative Biology and Pharmacology Studies. Sci Rep. 2018;8(1):4530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Sibley CP. Understanding placental nutrient transfer–why bother? New biomarkers of fetal growth. J Physiol. 2009;587(Pt 14):3431-3440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Blundell C, Tess ER, Schanzer AS, et al. A microphysiological model of the human placental barrier. Lab Chip. 2016;16(16):3065-3073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Costa MA. The endocrine function of human placenta: an overview. Reprod Biomed Online. 2016;32(1):14-43. [DOI] [PubMed] [Google Scholar]
- 139. Sastry BV. Techniques to study human placental transport. Adv Drug Deliv Rev. 1999;38(1):17-39. [DOI] [PubMed] [Google Scholar]
- 140. Liu F, Soares MJ, Audus KL. Permeability properties of monolayers of the human trophoblast cell line BeWo. Am J Physiol. 1997;273(5):C1596-C1604. [DOI] [PubMed] [Google Scholar]
- 141. Poulsen MS, Rytting E, Mose T, Knudsen LE. Modeling placental transport: correlation of in vitro BeWo cell permeability and ex vivo human placental perfusion. Toxicol in Vitro. 2009;23(7):1380-1386. [DOI] [PubMed] [Google Scholar]
- 142. Mathiesen L, Mose T, Mørck TJ, et al. Quality assessment of a placental perfusion protocol. Reprod Toxicol. 2010;30(1):138-146. [DOI] [PubMed] [Google Scholar]
- 143. Brandes JM, Tavoloni N, Potter BJ, Sarkozi L, Shepard MD, Berk PD. A new recycling technique for human placental cotyledon perfusion: application to studies of the fetomaternal transfer of glucose, inulin, and antipyrine. Am J Obstet Gynecol. 1983;146(7):800-806. [DOI] [PubMed] [Google Scholar]
- 144. Miller MT, Strömland K. Teratogen update: thalidomide: a review, with a focus on ocular findings and new potential uses. Teratology. 1999;60(5):306-321. [DOI] [PubMed] [Google Scholar]
- 145. Blundell C, Yi Y-S, Ma L, et al. Placental Drug Transport-on-a-Chip: A Microengineered In Vitro Model of Transporter-Mediated Drug Efflux in the Human Placental Barrier. Adv Healthc Mater. 2018;7(2):10.1002/adhm.201700786. doi: 10.1002/adhm.201700786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Miura S, Sato K, Kato-Negishi M, Teshima T, Takeuchi S. Fluid shear triggers microvilli formation via mechanosensitive activation of TRPV6. Nat Commun. 2015;6:8871. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Maliepaard M, Scheffer GL, Faneyte IF, et al. Subcellular localization and distribution of the breast cancer resistance protein transporter in normal human tissues. Cancer Res. 2001;61(8):3458-3464. [PubMed] [Google Scholar]
- 148. Kessler R. Engineered nanoparticles in consumer products: understanding a new ingredient. Environ Health Perspect. 2011;119(3):a120-a125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Esch MB, Mahler GJ, Stokol T, Shuler ML. Body-on-a-chip simulation with gastrointestinal tract and liver tissues suggests that ingested nanoparticles have the potential to cause liver injury. Lab Chip. 2014;14(16):3081-3092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Yin F, Zhu Y, Zhang M, Yu H, Chen W, Qin J. A 3D human placenta-on-a-chip model to probe nanoparticle exposure at the placental barrier. Toxicol In Vitro. 2019;54:105-113. [DOI] [PubMed] [Google Scholar]
- 151. Gude NM, Roberts CT, Kalionis B, King RG. Growth and function of the normal human placenta. Thromb Res. 2004;114(5-6):397-407. [DOI] [PubMed] [Google Scholar]
- 152. Rodriguez RJ. Management of respiratory distress syndrome: an update. Respir Care. 2003;48(3):279-86; discussion 286. [PubMed] [Google Scholar]
- 153. Fletcher K, Chapman R, Keene S. An overview of medical ECMO for neonates. Semin Perinatol. 2018;42(2):68-79. [DOI] [PubMed] [Google Scholar]
- 154. Coalson JJ. Pathology of new bronchopulmonary dysplasia. Semin Neonatol. 2003;8(1):73-81. [DOI] [PubMed] [Google Scholar]
- 155. Partridge EA, Davey MG, Hornick MA, et al. An extra-uterine system to physiologically support the extreme premature lamb. Nat Commun. 2017;8:15112. doi: 10.1038/ncomms15112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Labidi-Galy SI, Papp E, Hallberg D, et al. High grade serous ovarian carcinomas originate in the fallopian tube. Nat Commun. 2017;8(1):1093. doi:10.1038/s41467-017-00962-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7-30. [DOI] [PubMed] [Google Scholar]
- 158. Mehra KK, Chang MC, Folkins AK, et al. The impact of tissue block sampling on the detection of p53 signatures in fallopian tubes from women with BRCA 1 or 2 mutations (BRCA+) and controls. Mod Pathol. 2011;24(1):152-156. [DOI] [PubMed] [Google Scholar]
- 159. Meserve EEK, Brouwer J, Crum CP. Serous tubal intraepithelial neoplasia: the concept and its application. Mod Pathol. 2017;30(5):710-721. [DOI] [PubMed] [Google Scholar]
- 160. Soong TR, Howitt BE, Miron A, et al. Evidence for lineage continuity between early serous proliferations (ESPs) in the Fallopian tube and disseminated high-grade serous carcinomas. J Pathol. 2018;246(3):344-351. [DOI] [PubMed] [Google Scholar]
- 161. Dorayappan KDP, Gardner ML, Hisey CL, et al. A Microfluidic Chip Enables Isolation of Exosomes and Establishment of Their Protein Profiles and Associated Signaling Pathways in Ovarian Cancer. Cancer Res. 2019;79(13):3503-3513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Zhang P, Zhou X, He M, et al. Ultrasensitive detection of circulating exosomes with a 3D-nanopatterned microfluidic chip. Nat Biomed Eng. 2019;3(6):438-451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. de Almeida Monteiro Melo Ferraz M, Nagashima JB, Venzac B, Le Gac S, Songsasen N. A dog oviduct-on-a-chip model of serous tubal intraepithelial carcinoma. Sci Rep. 2020;10(1):1575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Turbeville HR, Sasser JM. Preeclampsia beyond pregnancy: long-term consequences for mother and child. Am J Physiol Renal Physiol. 2020;318(6):F1315-F1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Tranquilli AL, Dekker G, Magee L, et al. The classification, diagnosis and management of the hypertensive disorders of pregnancy: A revised statement from the ISSHP. Pregnancy Hypertens. 2014;4(2):97-104. [DOI] [PubMed] [Google Scholar]
- 166. Thomopoulos C, Tsioufis C, Michalopoulou H, Makris T, Papademetriou V, Stefanadis C. Assisted reproductive technology and pregnancy-related hypertensive complications: a systematic review. J Hum Hypertens. 2013;27(3):148-157. [DOI] [PubMed] [Google Scholar]
- 167. Opdahl S, Henningsen AA, Tiitinen A, et al. Risk of hypertensive disorders in pregnancies following assisted reproductive technology: a cohort study from the CoNARTaS group. Hum Reprod. 2015;30(7):1724-1731. [DOI] [PubMed] [Google Scholar]
- 168. Omani-Samani R, Alizadeh A, Almasi-Hashiani A, et al. Risk of preeclampsia following assisted reproductive technology: systematic review and meta-analysis of 72 cohort studies. J Matern Fetal Neonatal Med. 2020;33(16):2826-2840. [DOI] [PubMed] [Google Scholar]
- 169. Maynard S, Epstein FH, Karumanchi SA. Preeclampsia and angiogenic imbalance. Annu Rev Med. 2008;59:61-78. [DOI] [PubMed] [Google Scholar]
- 170. Trapiella-Alfonso L, Alexandre L, Fraichard C, et al. VEGF (Vascular Endothelial Growth Factor) Functionalized Magnetic Beads in a Microfluidic Device to Improve the Angiogenic Balance in Preeclampsia. Hypertension. 2019;74(1):145-153. [DOI] [PubMed] [Google Scholar]
- 171. Schug TT, Janesick A, Blumberg B, Heindel JJ. Endocrine disrupting chemicals and disease susceptibility. J Steroid Biochem Mol Biol. 2011;127(3-5):204-215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Diamanti-Kandarakis E, Bourguignon JP, Giudice LC, et al. Endocrine-disrupting chemicals: an Endocrine Society scientific statement. Endocr Rev. 2009;30(4):293-342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Hunt PA, Sathyanarayana S, Fowler PA, Trasande L. Female Reproductive Disorders, Diseases, and Costs of Exposure to Endocrine Disrupting Chemicals in the European Union. J Clin Endocrinol Metab. 2016;101(4):1562-1570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Jorgensen EM, Alderman MH 3rd, Taylor HS. Preferential epigenetic programming of estrogen response after in utero xenoestrogen (bisphenol-A) exposure. Faseb J. 2016;30(9):3194-3201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Acconcia F, Pallottini V, Marino M. Molecular Mechanisms of Action of BPA. Dose Response. 2015;13(4):1559325815610582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Stossi F, Bolt MJ, Ashcroft FJ, et al. Defining estrogenic mechanisms of bisphenol A analogs through high throughput microscopy-based contextual assays. Chem Biol. 2014;21(6):743-753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Bruno KA, Mathews JE, Yang AL, et al. BPA Alters Estrogen Receptor Expression in the Heart After Viral Infection Activating Cardiac Mast Cells and T Cells Leading to Perimyocarditis and Fibrosis. Front Endocrinol (Lausanne). 2019;10:598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Delfosse V, Grimaldi M, Pons JL, et al. Structural and mechanistic insights into bisphenols action provide guidelines for risk assessment and discovery of bisphenol A substitutes. Proc Natl Acad Sci U S A. 2012;109(37):14930-14935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Tang ZR, Zhang R, Lian ZX, Deng SL, Yu K. Estrogen-Receptor Expression and Function in Female Reproductive Disease. Cells. 2019;8(10):1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Newbold RR, Jefferson WN, Grissom SF, Padilla-Banks E, Snyder RJ, Lobenhofer EK. Developmental exposure to diethylstilbestrol alters uterine gene expression that may be associated with uterine neoplasia later in life. Mol Carcinog. 2007;46(9):783-796. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Duffy DC, McDonald JC, Schueller OJ, Whitesides GM. Rapid Prototyping of Microfluidic Systems in Poly(dimethylsiloxane). Anal Chem. 1998;70(23):4974-4984. [DOI] [PubMed] [Google Scholar]
- 182. Regehr KJ, Domenech M, Koepsel JT, et al. Biological implications of polydimethylsiloxane-based microfluidic cell culture. Lab Chip. 2009;9(15):2132-2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Toepke MW, Beebe DJ. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip. 2006;6(12):1484-1486. [DOI] [PubMed] [Google Scholar]
- 184. de Almeida Monteiro Melo Ferraz M, Henning HHW, Ferreira da Costa P, et al. Potential Health and Environmental Risks of Three-Dimensional Engineered Polymers. Environ Sci Technol Lett. 2018;5(2):80-85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185. Gokaltun A, Yarmush ML, Asatekin A, Usta OB. Recent advances in nonbiofouling PDMS surface modification strategies applicable to microfluidic technology. Technology. 2017;05(1):1-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Akther F, Yakob SB, Nguyen NT, Ta HT. Surface Modification Techniques for Endothelial Cell Seeding in PDMS Microfluidic Devices. Biosensors. 2020;10(11):182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Dickinson BL. UDEL® Polysulfone for Medical Applications. J Biomater Appl. 1988;3(4):605-634. [DOI] [PubMed] [Google Scholar]
- 188. Sastri VR. Chapter 8 - High-Temperature Engineering Thermoplastics: Polysulfones, Polyimides, Polysulfides, Polyketones, Liquid Crystalline Polymers, and Fluoropolymers. In: Sastri VR, ed. Plastics in Medical Devices Properties Requirements and Applications. 1st ed. Boston: William Andrew Publishing; 2010:175-215. [Google Scholar]
- 189. Ng SF, Rouse J, Sanderson D, Eccleston G. A Comparative Study of Transmembrane Diffusion and Permeation of Ibuprofen across Synthetic Membranes Using Franz Diffusion Cells. Pharmaceutics. 2010;2(2):209-223. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article because no data sets were generated or analyzed during the present study.