Skip to main content
Drug Metabolism and Disposition logoLink to Drug Metabolism and Disposition
. 2023 Sep;51(9):1157–1168. doi: 10.1124/dmd.123.001288

In Vitro and In Vivo Models for Drug Transport Across the Blood-Testis Barrier

Raymond K Hau 1, Stephen H Wright 1, Nathan J Cherrington 1,
PMCID: PMC10449102  PMID: 37258305

Abstract

The blood-testis barrier (BTB) is a selectively permeable membrane barrier formed by adjacent Sertoli cells (SCs) in the seminiferous tubules of the testes that develops intercellular junctional complexes to protect developing germ cells from external pressures. However, due to this inherent defense mechanism, the seminiferous tubule lumen can act as a pharmacological sanctuary site for latent viruses (e.g., Ebola, Zika) and cancers (e.g., leukemia). Therefore, it is critical to identify and evaluate BTB carrier-mediated drug delivery pathways to successfully treat these viruses and cancers. Many drugs are unable to effectively cross cell membranes without assistance from carrier proteins like transporters because they are large, polar, and often carry a charge at physiologic pH. SCs express transporters that selectively permit endogenous compounds, such as carnitine or nucleosides, across the BTB to support normal physiologic activity, although reproductive toxicants can also use these pathways, thereby circumventing the BTB. Certain xenobiotics, including select cancer therapeutics, antivirals, contraceptives, and environmental toxicants, are known to accumulate within the male genital tract and cause testicular toxicity; however, the transport pathways by which these compounds circumvent the BTB are largely unknown. Consequently, there is a need to identify the clinically relevant BTB transport pathways in in vitro and in vivo BTB models that recapitulate human pharmacokinetics and pharmacodynamics for these xenobiotics. This review summarizes the various in vitro and in vivo models of the BTB reported in the literature and highlights the strengths and weaknesses of certain models for drug disposition studies.

SIGNIFICANCE STATEMENT

Drug disposition to the testes is influenced by the physical, physiological, and immunological components of the blood-testis barrier (BTB). But many compounds are known to cross the BTB by transporters, resulting in pharmacological and/or toxicological effects in the testes. Therefore, models that assess drug transport across the human BTB must adequately account for these confounding factors. This review identifies and discusses the benefits and limitations of various in vitro and in vivo BTB models for preclinical drug disposition studies.

Introduction

The blood-testis barrier (BTB) is a critical component of the male reproductive tract that safeguards the normal spermatogenic process from external threats. It is a selectively permeable cell barrier formed by adjacent Sertoli cells (SCs) in the seminiferous tubules that express intercellular junctional proteins to limit the accumulation of reproductive toxicants in the male genital tract (Dym and Fawcett, 1970). Except for spermatogonia, all developing germ cells are protected within the adluminal compartment of the seminiferous tubules. In addition to the network of membranes that physically blocks access to the male genital tract, SCs also express various transporters along their basal membranes to regulate the disposition of compounds across the BTB (Mital et al., 2011; Mruk et al., 2011; Mruk and Cheng, 2015). Moreover, SCs express and secrete immunomodulatory factors to achieve testicular immune privilege (Fijak and Meinhardt, 2006; Li et al., 2012; Zhao et al., 2014). Together, these three factors actively protect developing germ cells residing on either side of the BTB. However, the seminiferous tubule lumen can act as a sanctuary site for some viruses (e.g., Ebola, HIV, Zika) and cancers (e.g., leukemias) because certain xenobiotics cannot effectively cross the tight SC epithelia to achieve pharmacologically relevant concentrations in the adluminal compartment. There are many reports of testicular persistence and/or relapse of viruses and cancers, despite eradication from the blood and other tissues (Nesbit et al., 1980; Ritzén, 1990; Byrn and Kiessling, 1998; Kiessling et al., 1998; Politch et al., 2012; Houzet et al., 2014; Deen et al., 2017; Robinson et al., 2018; Ma et al., 2021). This phenomenon highlights a need to identify the mechanisms that permit antiviral or antineoplastic therapies to successfully treat these diseases in the testes.

Uptake transporters present at the basal membrane of SCs are vital for the delivery of small molecule therapeutics across the BTB. Pharmacologically relevant uptake transporters include the organic cation transporters (OCTs), novel organic cation transporters (OCTNs), organic anion transporters (OATs), organic anion transporting polypeptides (OATPs), and equilibrative nucleoside transporters (ENTs). Efflux transporters, such as P-glycoprotein (P-gp), breast cancer resistance protein (BCRP), and the multidrug resistance-associated proteins (MRPs), act in opposition to prevent the accumulation of toxic molecules in SCs, although not all uptake transporter substrates are (good) efflux transporter substrates. There are also dozens of other understudied BTB transporters that can influence the disposition of drugs to treat testicular indications, although they will not be discussed in this review. Collectively, the listed transporters are involved in the flux of many clinically relevant drugs and environmental toxicants, but many of these BTB pathways have not been adequately defined in rodents, humans, and other species.

There are in vitro and in vivo BTB models across different species reported in the literature, but there is a significant lack of information on their relevance for drug transport studies. As a result, there is a need to evaluate the benefits and limitations of these in vitro and in vivo BTB models to assess testicular drug disposition. Furthermore, the information that is known about some efflux transport pathways, such as MRP4 and BCRP, indicate that rodent models may not recapitulate events in humans (Hau et al., 2021, 2023). Therefore, a standardized array of clinically relevant BTB models and assays to study drug disposition across the BTB is warranted. The purpose of this review is to provide a comprehensive overview of the mammalian BTB models described in the literature and to outline their potential in preclinical drug disposition studies.

In Vitro Models

Rodent Sertoli Cell Models

Rodent SC models are commonly used for a variety of applications to study the physiology of the testes and the BTB. This is due to the comparatively easy access to fresh rodent testes for cell isolation and the mandatory use of rodent models in preclinical studies for assessing reproductive toxicity. Historically, BTB drug transport has been studied with primary SCs isolated from immature and mature rats or mice. Immature rodents are used due to their ease of isolation and the lack of contaminating germ cells for SC isolation; however, mature rodents with an established BTB are more physiologically relevant. In vivo, the establishment of the functional and complete BTB occurs after 15 to 18 days postpartum in mice (Moroi et al., 1998; Hogarth, 2015; Willems et al., 2010; Xiong et al., 2018) and 15 to 21 days postpartum in rats (Bergmann and Dierichs, 1983; Russell et al., 1989; Toyama et al., 2001). This is important because data collected using immature SCs may not be representative of the findings collected in adult animals due to ontogenetic changes in transporter expression (Brouwer et al., 2015; Cheung et al., 2019; van Groen et al., 2021; Felmlee and Zhang, 2022). These transporter expression changes may be, in part, associated with changing hormone or immunomodulatory factor levels as animals mature and spermatogenesis begins. Interestingly, one study observed downregulation of P-gp in rat SCs after exposure to estradiol-17β, testosterone, and transforming growth factor-β3 but no change after exposure to interleukin-1α and tumor necrosis factor-α (Su et al., 2012). On the other hand, estradiol-17β and interleukin-1α also upregulated rat OATP1A5, but the other hormones or cytokines had no effect. Hormone and cytokine levels can fluctuate significantly over time and are also sensitive to various environmental factors (e.g., infections) (Larsson et al., 2015; Ter Horst et al., 2016; Bell, 2018). For example, testosterone levels are generally low after birth and gradually rise during peripuberty due to the onset of spermatogenesis, but its downregulatory effect on P-gp is counterintuitive. It is possible that hormone and cytokine levels indirectly regulate the intracellular levels of various biomolecules (e.g., steroids, prostaglandins, and nucleosides) by modulating transporter expression. However, it is clear that the effect of hormones and cytokines on transporter expression warrants further evaluation because there are dozens of other physiologically and/or pharmacologically relevant transporters expressed in the testes.

It is also possible that transporter expression and localization can be altered during exposures to certain environmental toxicants, such as zearalenone or cadmium (Su et al., 2012; Koraïchi et al., 2013). Cadmium chloride exposure was shown to downregulate P-gp, MRP1, ABCG1, OATP1A5, PEPT1, and ZIP8 in cultured rat SCs, whereas neonatal exposure to zearalenone causes MRP4 to redistribute from the basal membrane to the apicolateral membrane of SCs. It is known that certain disease states affect transporter expression in other tissues (Evers et al., 2018). Therefore, it is conceivable that some testicular indications may cause changes in transporter expression and, ultimately, affect the disposition of drugs to the testes. Thus, it is critical to account for age, environmental exposures, and other confounding factors when assessing in vitro and in vivo drug disposition to the testes. This process may involve the use of more than one in vitro and in vivo model and/or the use of multiple animal replicates, but it is necessary to make an informed conclusion.

While drug transport is often performed on flat monolayers of nonpolarized cells plated on standard plastic cell culture vessels, differences in transporter expression between immature and mature SCs can affect drug disposition across these membranes. Additionally, drug transport on these nonpolarized monolayers is not representative of in vivo epithelial barriers due to cell polarization directing transporter localization (Cao et al., 2012). Junctional complexes formed between adjacent SCs separate the cell membrane into apical/apicolateral or basal/basolateral domains, leading to differential transporter expression on each side. There is no distinction between the basal and apicolateral transporters when SCs are cultured as flat monolayers. Consequently, it is possible that while transporter-mediated uptake of a compound may be observed in vitro, it would not cross the in vivo BTB because the transporter is expressed at the apicolateral membrane of SCs. Transporter expression and localization maps such as the one described by Hau et al. (Hau et al., 2023) for human testes could provide some context, but functional studies to validate these findings are still warranted.

The specificity, selectivity, expression, and localization of drug transporters at the BTB can vary between species (Chu et al., 2013; Hau et al., 2021). For example, a previous study showed that, whereas MRP4 localizes to the basal membrane of SCs in humans and rhesus macaques, expression is restricted to the apicolateral membrane of SCs in normal rodents (Klein et al., 2014). Several other studies reported basal membrane localization of MRP4 in humans (Morgan et al., 2012; Huang et al., 2016). Importantly, another study observed MRP4 at the basal membrane of SCs, Leydig cells, and peritubular myoid cells in rats but found that it redistributes to the apicolateral membrane of SCs, spermatocytes, and spermatids after neonatal treatment with zearalenone (Koraïchi et al., 2013). Interestingly, unlike in rats or humans, MRP4 in mice is exclusively localized to the Leydig cells (Morgan et al., 2012, 2015). These contradictory findings are not limited to MRP4, as they have been observed with another major efflux transporter, BCRP. In humans, BCRP is localized to the basal and apicolateral membranes of SCs, peritubular myoid cells, developing germ cells, and vascular endothelial cells (Melaine et al., 2002; Huang et al., 2016). Notably, although BCRP in rats is expressed in the same cells as humans, it is absent at the basal membrane of SCs (Qian et al., 2013). Furthermore, BCRP is exclusively localized to the vascular endothelial cells in mouse testes (Enokizono et al., 2007). These observations suggest that the use of rodent models for studying drug transport across the BTB may not always be representative of what is observed in humans. Nevertheless, studying drug transport with these in vitro rodent models can provide insight into at least some of the transporter mechanisms present in SCs across species. Indeed, flat monolayers of primary rodent SCs have been used to meticulously study the interaction between nucleosides and nucleoside analogs with the ENTs (Kato et al., 2005, 2006, 2009; Klein et al., 2013; Miller et al., 2021a). Unfortunately, this model only describes basic drug-transporter interactions and does not account for transporter interactions at specific membranes of a polarized SC.

Although these basic rodent monolayer models are a valuable tool, the development of a two-compartment cell culture system to study transepithelial permeability and polarized chemical secretion across primary rodent SCs was a significant breakthrough (Janecki and Steinberger, 1986, 1987; Janecki et al., 1987; Steinberger and Klinefelter, 1993). These polarized cell models are much more representative of the in vivo BTB; however, they still neglect the potential physiologic importance of the peritubular myoid cells and developing germ cells that, along with the SCs, compose the seminiferous tubules. Initial studies with these primary rat SC layers reported transepithelial electrical resistance (TEER) values exceeding 100 Ohms•cm2 (Janecki et al., 1991a,b; Steinberger and Klinefelter, 1993). More recent studies observed peak TEER values of 70 Ohms•cm2 from primary rat SCs (Kaitu’u-Lino et al., 2007; Nicholls et al., 2009; Mruk and Cheng, 2011). Similarly, TEER values in primary mouse SCs are reported to reach up to 60 Ohms•cm2 (Wu et al., 2017a,b; Hu et al., 2021; Liu et al., 2021). But these are comparatively low TEER values compared with those observed in Caco-2 and MDCK cells, which surpass 1000 Ohms•cm2 when the tight junctions are fully developed (Prozialeck and Lamar, 1997; Srinivasan et al., 2015). It is possible that the unusual morphology and formation of tight junctions near the basal membrane of SCs contribute to the lack of a coherent, 3D epithelial monolayer on two compartment cell culture inserts. As a result, SCs plated in two-compartment inserts may permit paracellular diffusion of some chemicals that would not occur in vivo and, consequently, cannot accurately reflect the significantly tighter in vivo BTB. Although there is currently no way to directly measure the TEER of an intact epithelial barrier in vivo, a recent study showed that the membrane-impermeable protein biotinylation reagent sulfo-NHS-LC-biotin was unable to penetrate the intact in vivo BTB but was unimpeded when the barrier was disrupted by CdCl2 (Chen et al., 2018). Furthermore, other studies have demonstrated that the intact in vivo BTB was impenetrable by various tracers including lanthanum and NHS-linked biotin (Neaves, 1973; Tarulli et al., 2008; Carette et al., 2010; McCabe et al., 2010; Pelletier, 2011). One study assessed the membrane permeability of primary mouse SCs; however, it was not relevant to the paracellular permeability of the BTB (Liu et al., 2020). These findings suggest that the disposition of some small molecules across the BTB is largely impossible without the help of carrier-mediated processes like transporters.

Previous studies have assessed the transepithelial transport of [3H]L-carnitine and [14C]TEA across primary rat SCs cultured on two-compartment inserts (Kobayashi et al., 2005; Maeda et al., 2007). Based on higher transporter affinity for L-carnitine, compared with TEA, OCTN2 was suggested to be responsible and to localize to the basal membrane of rat SCs and OCTN1 to the apicolateral membrane. L-carnitine is critical for maintaining normal testicular physiology to support spermatogenesis and is the endogenous substrate of OCTN2 (Tamai et al., 1998; Wang et al., 1999; Palmero et al., 2000; Agarwal and Said, 2004; Mongioi et al., 2016). Furthermore, OCTN2 is known to localize to the basal membrane of human SCs, where it may support the uptake of L-carnitine and other molecules across the BTB (Hau et al., 2022a, 2023). A similar study that evaluated transepithelial transport of [3H]adjudin across primary rat SCs demonstrated that it is a substrate for OCTN2, OATP1A5, OATP6B1, and OATP6C1 expressed at the basal membrane (Su et al., 2011). But, as described previously, these transepithelial transport experiments are not adequate models of the in vivo BTB due to the absence of tighter intercellular junctional complexes to prohibit paracellular diffusion and the lack of other testicular cells that can influence SC protein expression. For example, developing germ cells are known to stimulate the expression of genes such as transferrin in SCs (Djakiew and Dym, 1988; Le Magueresse et al., 1988; Stallard and Griswold, 1990). Furthermore, the peritubular myoid cells that interact with SCs are also partly responsible for generating and maintaining the extracellular matrices that affect junctional protein dynamics at the BTB (Skinner et al., 1985). Consequently, a simplified in vitro transepithelial transport model of the rodent BTB is insufficient to reproduce the complexities of the in vivo barrier. Similarly, transport assays with nonpolarized monolayers of rodent SCs can provide higher throughput compared with two-compartment models, but, as noted earlier, the absence of normal epithelial structure compromises the interpretation of the results. Nevertheless, despite these limitations, and with important caveats outlined here, drug transport assays conducted with primary rodent SCs cultured as a monolayer on standard cell cultureware or two-compartment inserts are important for exploratory in vitro studies.

To expedite studies on the male reproductive system, several immortalized rodent SC lines have been established. One of the most used rodent SC lines are TM4 cells, which were isolated from the testis of 11- to 13-day-old, immature BALB/c mice (Table 1) (Mather, 1980). Since these cells were isolated from an immature mouse, drug transport data generated from TM4 cells may not be accurately reflected in studies with mature SCs or animals. Nevertheless, TM4 cells have been used to study a variety of basic physiologic functions in the testis including cell signaling, metabolism, compound transport, and BTB integrity. Basic transport studies have been performed with flat monolayers of TM4 cells plated on standard plastic cell culture plates (Selva et al., 2004; Kato et al., 2005, 2009; Robillard et al., 2012; Huang et al., 2014; Hoque et al., 2015; Ge et al., 2018; Kubo et al., 2022). Studies assessing the transport of clinically relevant drugs in TM4 cells have been largely limited to nucleoside-based antivirals (Kato et al., 2005, 2009; Robillard et al., 2012; Hoque et al., 2015). However, there are many transporters that can transport other clinically relevant compounds across SCs that have not been analyzed in TM4 cells. Compared with primary rat SCs, TM4 cells had a lower capacity for uridine uptake over 10 minutes and were mediated by a saturable and nonsaturable component (Kato et al., 2005). TM4 cells also had negligible functional expression of the Na+-dependent concentrative nucleoside transporters, whereas the concentrative nucleoside transporters appeared to mediate at least 25% of the uridine transport in primary rat SCs (Kato et al., 2005). Furthermore, the affinity of various endogenous nucleosides and nucleobases appeared to vary between primary rat SCs and TM4 cells, which may be attributed to species differences in transporter expression and function (Kato et al., 2005, 2006, 2009). These observations differ when compared with primary human SCs and human TERT-immortalized SC (hT-SerC) line, which appear to only functionally express ENT1 (Hau et al., 2020).

TABLE 1.

List of immortalized rodent SCs published in the literature

Cell Line Species (Strain) Age References
15P-1 Mus musculus (C57BL/6J x DBA/2 PyLT transgenic) 6 months postpartum Paquis-Flucklinger et al., 1993
42GPA9 Mus musculus (Tg(PyLT) transgenic) 6 months postpartum Bourdon et al., 1998
93RS2 Rattus norvegicus (CD) 16 days postpartum Jiang et al., 1997
A3/C2# Mus musculus (C57BL/6J) 6 weeks postpartum Sato et al., 2013
ASC-17D Rattus norvegicus (Sprague-Dawley) 120 days postpartum Roberts et al., 1995
C3H Mus musculus (C3H Heston) 16 months postpartum Franks, 1968
G5/SG5/J5# Mus musculus (BALB/c) 6 days postpartum Hofmann et al., 2003
MSC-1 Mus musculus (hMIS-SV40 transgenic) 13 weeks postpartum Peschon et al., 1992
PASC1 Rattus norvegicus (Sprague-Dawley) adult* Kabbesh et al., 2021
RTS3-3 Rattus norvegicus (Wistar SV40-tsA58 transgenic) adult* Tabuchi et al., 2003
S14-1 Mus musculus (BALB/c) 20 days postpartum Boekelheide et al., 1993
SCIT-C8 Rattus norvegicus (Wistar) 19 days postpartum Konrad et al., 2005
SerW3 Rattus norvegicus (Wistar) 17 days postpartum Pognan et al., 1997
SMAT1 Mus musculus (B6.D2 x B6.CBA F2 AMH-SV40 transgenic) 6.5 days postpartum Dutertre et al., 1997
TM4 Mus musculus (BALB/c) 11–13 days postpartum Mather, 1980
TR-ST Rattus norvegicus (Long-Evans) 2 years postpartum Mather et al., 1982
TTE3 Mus musculus (C57BL/6J SV40-tsA58 transgenic) 8 weeks postpartum Tabuchi et al., 2002

#Multiple clones were generated in the study.

*Age was not reported at the time of cell isolation.

In addition to drug transport across flat monolayers, a few studies have assessed transepithelial transport across TM4 cells in a two-compartment system, but there are conflicting reports on TEER values for unperturbed TM4 cells. Whereas some studies report TEER values of ∼20 to 60 Ohms•cm2 (Papadopoulos et al., 2016; Schrade et al., 2016; Wang et al., 2022; Zhang et al., 2022), others report significantly higher values (> 200 Ohms•cm2) (Hu et al., 2014; Zhou et al., 2018; Fisher et al., 2019). These conflicting values may be attributed to different culture systems and methods (note: some publications incorrectly reported TEER as Ohms/cm2 rather than Ohms•cm2, and these values have been recalculated here based on available information). While transepithelial transport studies may be adequately performed with TM4 cells achieving TEER values of > 200 Ohms•cm2, the reproducibility and clinical relevance is questionable due to the TEER values observed in primary mouse SCs (Wu et al., 2017a,b; Hu et al., 2021; Liu et al., 2021) and differing expression, localization, and selectivity/kinetics of the mouse transporter orthologs compared with other species (Hau et al., 2022a, 2021, 2023).

There are many other immortalized rodent (rat or mouse) SC lines, albeit generated from animals of different ages, and these are listed in Table 1. Unlike primary rodent SCs or TM4 cells, fewer studies assessing transporter function have been performed with these cell lines. One study investigated the function of the H+-coupled monocarboxylate transporter 4 on lactate transport in 15P-1 cells following cadmium or lead exposure (Yu et al., 2019). Monocarboxylate transporter 4 predominantly exports lactate out of cells, although it can function as a bidirectional transporter for other endogenous monocarboxylates and interacts with monocarboxylate xenobiotics like indomethacin and diclofenac (Manning Fox et al., 2000; Sasaki et al., 2016; Contreras-Baeza et al., 2019). Nevertheless, the utility of mouse 15P-1 cells when directly investigating xenobiotic disposition remains understudied. Similarly, there is only one published report on transporters in mouse 42GPA9 cells; however, this study was limited to L-ascorbic acid transporters (Angulo et al., 2008). There have been several studies assessing the reproductive toxicity of dichlorodiphenyltrichloroethane, doxorubicin, and other toxicants in the SerW3 cell line (Fiorini et al., 2004; Bernard et al., 2007; Fiorini et al., 2008; Qiu et al., 2016; Tremblay and Delbes, 2018), although there are only two additional studies that were limited to investigating the interaction between flusilazole or zearalenone with ATP-binding cassette transporters (Koraïchi et al., 2013; Karacaoğlu, 2022). Unfortunately, the expression and function of pharmacologically-relevant uptake transporters have not been explored in any of these immortalized rodent cell lines, which is a major disadvantage for their informed use in drug disposition studies.

The use of rodent BTB models carries an important caveat; namely, they are not always representative of clinical findings. Some transporters, such as rodent and human OCT1, share similar selectivity and kinetics for at least some drugs; however, other OCT1 substrates/inhibitors may diverge from this trend (Floerl et al., 2020). Additionally, functional differences in P-gp transport between species have been shown to affect drug disposition across the blood-brain barrier (Syvanen et al., 2009). Consequently, assessing drug disposition across the BTB in different species may be subject to similar correlation issues. Drug transport with primary rodent SCs is straightforward because gene expression in immortalized rodent SC lines can be highly variable and fresh primary cell isolation is convenient. However, primary rodent SCs may not express orthologous transporters to human cells, and the selectivity, kinetics, and localization of the orthologous transporters that are expressed can be significantly different (Chu et al., 2013; Hau et al., 2021, 2023). Furthermore, the availability of human SC models and other heterologous expression systems for human transporters renders in vitro rodent models less useful. In cases where in vitro rodent models are used, it is extremely important to validate the generated drug transport data with more clinically relevant models for testicular drug disposition.

Human Sertoli Cell Models

Unlike rodent SCs, obtaining freshly isolated, healthy human SCs is difficult due to the relative rarity of healthy donor tissue and the associated regulatory and logistical hurdles. Furthermore, the human BTB is not completely established until puberty (around age 12–13), reflecting the absence of developing germ cells that require the protection afforded by the BTB (Furuya et al., 1978). This corresponds to observations made in the testes of rodents, where the BTB is not completely formed until later in life. Consequently, it is fundamentally important to use adult human SCs for BTB studies instead of those from a fetus or child due to major differences in gene expression, physiology, and morphology. Isolation of primary adult human SCs has been reported in many studies (Lipshultz et al., 1982; Holmes et al., 1983; Buch et al., 1988; Berensztein et al., 1992; Santiemma et al., 1992; Cudicini et al., 1997; Chandley et al., 1996; Cowan et al., 2010; He et al., 2010; Chui et al., 2011; Guo et al., 2015; Jesus et al., 2016; Wen et al., 2017), but the modest yield of these preparations limits the number of studies that can be conducted at one time. Gene expression profiles in primary cells are generally unstable after dissociation from the native tissue and these cells will eventually undergo cellular senescence (Hayflick, 1965; Olovnikov, 1996). Because of these practical difficulties and limitations associated with the use of primary human SCs, heterologous expression systems and/or nonhuman SCs have been the gold standard for drug transport studies.

Although there are many published reports recognizing the importance of drug transport at the BTB, there are few studies that directly assess the function of pharmacologically relevant drug transporters in human SCs (Klein et al., 2013; Hau et al., 2020; Miller et al., 2021a; Hau et al., 2022b). Many of these studies have primarily focused on the function of Na+-independent ENT1 and ENT2. Other common uptake and efflux transporters are expressed in human testes (Kullak-Ublick et al., 1995; Melaine et al., 2002; Pizzagalli et al., 2002; Suzuki et al., 2003; Bart et al., 2004; Bleasby et al., 2006; Huber et al., 2007; Klaassen and Aleksunes, 2010; Robillard et al., 2012; Fietz et al., 2013; Klein et al., 2014; Huang et al., 2016; Hau et al., 2022a), but little is known about how these other transporters collectively affect drug disposition across the BTB. Due to the importance of identifying reproductive toxicity during drug development, understanding transporter-mediated disposition across the human BTB and which models are appropriate is vital for successful preclinical and clinical studies.

Currently, there are two immortalized human SC lines that have been reported in the literature (Wen et al., 2017; Hau et al., 2020). Both cell lines were generated by transducing human telomerase reverse transcriptase from the primary SCs of adult patients. The hS1 cell line was generated from the primary SCs of an adult male of unknown age with obstructive azoospermia (Wen et al., 2017). However, these cells are not publicly available, and drug transport studies with them have not been reported. Conversely, the mRNA expression of common drug transporters was described in hT-SerCs, which were derived from a recently deceased, healthy 36-year-old adult male (Chui et al., 2011; Gaur et al., 2018; Hau et al., 2020). Functional transport studies with hT-SerCs have been reported, although the protein expression levels of many transporters are unknown (Hau et al., 2020; Miller et al., 2021a; Hau et al., 2022b). The novel male contraceptives, H2-gamendazole and gamendazole, are known to exert reversible infertility in several species and are taken up by human and rat SCs by one or more transporters, given its large structure and negative charge at physiologic pH (Tash et al., 2008a; Tash et al., 2008b; Gupta et al., 2011; Hau et al., 2022b). It is speculated that the transporter(s) responsible for H2-gamendazole uptake is (are) shared between species; however, additional work is necessary to identify each specific transporter. However, species differences in testicular transporter expression, localization, and function have been reported and is a major concern for drug disposition to the testes (Melaine et al., 2002; Enokizono et al., 2007; Morgan et al., 2012; Klein et al., 2013; Koraïchi et al., 2013; Klein et al., 2014; Morgan et al., 2015; Huang et al., 2016; Hau et al., 2020, 2021, 2023).

It is important to note that these transport studies were conducted using monolayers of hT-SerCs on standard plastic cell culture plates. Unfortunately, hT-SerCs and primary human SCs do not form a tight, polarized epithelial monolayer on two-compartment systems. Compared with rodent SCs, MDCK cells, and Caco-2 cells, human SCs form a leakier in vitro barrier, as is evident by low TEER values ranging from ∼10 to 30 Ohms•cm2 (Chui et al., 2011; Chen et al., 2017; Siemann et al., 2017; Gaur et al., 2018; Hau et al., 2020). Consequently, human SCs cultured on two-compartment inserts permit significant paracellular diffusion of test compounds, and transporter-mediated transepithelial flux of these compounds is difficult to resolve above this background signal. Overexpression of tight junction proteins in hT-SerCs to promote intercellular interactions has been attempted (unpublished data); however, there was no difference in TEER values compared with untransfected cells. This limits their utility and relevance in preclinical drug disposition studies; however, like rodent SCs, human SCs are one of many predictive tools to study drug transport across the BTB. Human SCs may be a better predictive tool of drug disposition across the BTB since they express human transporters that may exhibit differing substrate selectivity or kinetics, although further work is needed to provide a more comprehensive comparison.

Other Mammalian Sertoli Cell Models

Although isolation of SCs from other mammalian species is feasible and has been done for pigs, bulls, sheep, canines, nonhuman primates, and other animals (Vazeille and Chevalier, 1980; De Martino et al., 1985; Renier et al., 1986; Tung et al., 1987; Coombs and Jenkins, 1988; Monet-Kuntz and Fontaine, 1989; Merhi et al., 2001; Davidson et al., 2007; Guibert et al., 2011; Zhang et al., 2013), the clinical relevance of these models is ambiguous. In addition, functional drug transporter studies have not been performed with any of these mammalian SCs. Although there have been reports on BTB transporter expression and localization in nonhuman primates and horses (Klein et al., 2014; Merkl et al., 2016), it is restricted to only the BTB localization of MRP1, MRP4, MRP5, and MRP8 in mature rhesus macaques and to the ATP-binding cassette transporter A1 in horses. The expression and localization of common pharmacologically relevant uptake transporters at the BTB are unknown in these species; however, nonhuman primate models are generally regarded as the closest representation of human physiology. An extensive map of transporter localization, selectivity, and kinetics for the relevant species must be generated first before widespread adoption of these other models in BTB drug transport studies can ensue. Currently, the use of primary human SCs, SC lines, or heterologous expression systems for simple drug transporter studies across the BTB is preferable to cells from other mammalian species.

Complex 3D Culture Models

Although current SC in vitro models are largely limited to conventional flat monolayers of cells plated on standard cell cultureware or on two-compartment inserts, some studies have used co-culture methods to improve the genotypical or phenotypical characteristics of two-compartment cell systems (Hilgendorf et al., 2000; Megard et al., 2002; Zhang et al., 2011). However, reconstructing a tight SC barrier to study active transport across the BTB remains a major objective. As previously explained, SCs tend to form weak intercellular interactions in vitro on two-compartment inserts, even with cell culture additives, matrices, and co-cultures. Furthermore, assessing transepithelial transport of, or inhibition of transport by, tens or hundreds of different compounds with these systems is unrealistic. Most compounds interact with more than one transporter and the transporters expressed by peritubular myoid cells and interstitial Leydig cells may affect drug disposition across the BTB, since some of the same common drug transporters are also expressed by these cells as in SCs (Klein et al., 2014; Hau et al., 2022a, 2023). Therefore, more complex in vitro BTB models that more effectively recapitulate in vivo physiology are warranted.

Testicular organoids, ex vivo models, and other 3D cultures have gained substantial popularity in recent years due to their closer approximation to in vivo physiology. Many advanced testicular models have been reported in the literature with different approaches to generate a more physiologically relevant model, as reported in Table 2. However, some of these systems are derived from fetal tissues and may be less representative of an established BTB in adult animals. Furthermore, some of these models rely on extracellular matrices to maintain the desired cellular aggregate morphology, which can adversely affect drug transport studies. Although some of these complex models are also grown on two-compartment inserts, they aggregate and adopt a spheroid-like morphology unlike the flat monolayers in the previously described models. Drug transporter function has also been studied in organoid models, but these experiments rely on fluorescent substrates that are not available for some transporters (Achilli et al., 2014; Zhang et al., 2019). Recently, one study demonstrated that measuring transport of radiolabeled glucose, fructose, or dipeptide glycyl-sarcosin across human intestinal organoids was feasible (Zietek et al., 2020). Unfortunately, these techniques have not been applied to testicular organoid models. An ex vivo assay to assess radiolabeled uridine transport across intact rat seminiferous tubules has been described (Klein et al., 2013), but this method is technically challenging, time-intensive, and does not adequately define the role of apicolateral transporters in SC transcellular transport, which may be important for the disposition of certain compounds into the adluminal compartment of the seminiferous tubule. It is feasible that drug transport assays can be adapted for use in some of the cultured models listed in Table 2, but they would need to be individually optimized for radiolabeled or fluorescent substrates and for unlabeled compounds detected by liquid chromatography-tandem mass spectrometry. It is important to note that these complex models can be used for other applications when studying reproductive physiology, but high-throughput drug transport studies with these models are impractical and are best suited for the most promising drug candidates.

TABLE 2.

List of complex rodent or human 3D culture models that may be adapted for use in low-throughput drug transport studies

Model Species Age References
Air-liquid interface Mus musculus (male pCXN-GFP, Acr-GFP, or Gsg2-GFP transgenic x female ICR, C57BL/6J, or ICR x C57BL/6F1 F2) 0.5–14.5 days postpartum Gohbara et al., 2010; Komeya et al., 2016; Sato et al., 2011a; Sato et al., 2011b; Yokonishi et al., 2013; Yokonishi et al., 2014
Mus musculus (Acr-GFP, Gsg2-GFP, and WBB6F1-Sl/Sld transgenic) 2.5 days–28 weeks postpartum Sato et al., 2015
Mus musculus (CD-1) 2.5–37 days postpartum Arkoun et al., 2015; Dumont et al., 2015; Dumont et al., 2016; Rondanino et al., 2017
Mus musculus (C57BL/6J or CBAB6F1) 3–5 days postpartum Reda et al., 2017; Richer et al., 2021
Mus musculus (ddY) 5 days postpartum Suzuki and Sato, 2003
Rattus norvegicus (Sprague-Dawley) 5–7 days postpartum Liu et al., 2016; Reda et al., 2016
Rattus norvegicus (Long-Evans) 12–14 days postpartum Steinberger and Steinberger, 1965; Steinberger et al., 1964
Homo sapiens 6–12 weeks gestation fetus Lambrot et al., 2006
2–12 years prepubertal de Michele et al., 2017
77.94 ± 6.84 years Roulet et al., 2006
Adult* Baert et al., 2020; Vermeulen et al., 2018
Hanging drop Mus musculus (CD-1) Gestation fetus* (adult females aged 2–3 months) Potter and DeFalco, 2015
Homo sapiens 7–9 weeks gestation fetus Jørgensen et al., 2015
Adult* Jørgensen et al., 2014; Vermeulen et al., 2018
Microfluidic device Mus musculus (male Acr-GFP transgenic x female ICR or B6D2F1 F2) 0.5–5.5 days postpartum Komeya et al., 2016
Mus musculus (CD-1) 7–8 days postpartum AbuMadighem et al., 2022
Organoid-based cellular aggregate or seminiferous tubule cord (grown on different extracellular matrices) Mus musculus (unknown strain) 8–10 days postpartum Sakib et al., 2019
Mus musculus (C57BL/6J) 6 days postpartum Zhang et al., 2014
Mus musculus (BALB/c) 1 2, 4, or 8 weeks postpartum Abu Elhija et al., 2012
Mus musculus (CD-1) 5 days–16 weeks postpartum AbuMadighem et al., 2018; Edmonds and Woodruff, 2020; Enders et al., 1986; Stukenborg et al., 2008
Rattus norvegicus (Sprague-Dawley) 5–60 days postpartum Alves-Lopes et al., 2017; Hadley et al., 1985; Hadley et al., 1990; Harris et al., 2015; Harris et al., 2016; Lee et al., 2006b; Lee et al., 2011; Legendre et al., 2010; Reda et al., 2014; Reuter et al., 2014; Zhang et al., 2017
Rattus norvegicus (CD) 7 days postpartum Gassei et al., 2008; Gassei et al., 2010; Pan et al., 2013
Rattus norvegicus (SIV-1) Newborn, 8–10, 18–25, 35–40, and 90 days postpartum Zenzes and Engel, 1981
Rattus norvegicus (unknown albino strain) 25 days postpartum Enders et al., 1986
Homo sapiens 5–9.5 weeks gestation fetus Oliver et al., 2021
6 months–75 years Baert et al., 2017; Gholami et al., 2018; Jabari et al., 2020; Mincheva et al., 2018; Pendergraft et al., 2017; Sakib et al., 2019; von Kopylow et al., 2018
Adult* Lee et al., 2006a; Lee et al., 2007
Whole seminiferous tubule Rattus norvegicus (Sprague-Dawley) 8 days–15 weeks postpartum Klein et al., 2013; Mao et al., 2021; Perrard et al., 2016
Rattus norvegicus (Brown Norway) 3–5 months postpartum Li et al., 2016
Homo sapiens 25–83 years Perrard et al., 2016; Seidl and Holstein, 1990

*Age was not reported at the time of isolation.

Nevertheless, the adaptation of these technologies and techniques for measuring transport of select compounds across complex testicular culture models is promising. The widespread use of these models is still limited by cost, time, and throughput compared with conventional models. Furthermore, the use of heterologous expression systems to study the selectivity and kinetics of one or more human transporters may be appropriate if the transporter(s) that interact with a drug of interest have been defined. Transport in heterologous expression systems is the same as transport in SCs, although some drugs can interact with multiple efflux transporters expressed by SCs that may not be present in the former model. Despite these limitations, the information gained through each of these in vitro models (e.g., primary/immortalized rodent and human SC monolayers, heterologous expression systems, complex 3D culture models) for BTB drug disposition is highly valuable prior to applying it to in vivo models.

In Vivo Models

Rodent Models

Current U.S. Food and Drug Administration (FDA) guidelines recommend the use of a rodent or nonrodent animal model to assess the pharmacokinetic and toxicokinetic characteristics of an investigational new drug (https://www.fda.gov/regulatory-information/search-fda-guidance-documents/m3r2-nonclinical-safety-studies-conduct-human-clinical-trials-and-marketing-authorization). The use of rats and mice, among other preclinical species, for assessing testicular disposition of xenobiotics is standard practice in biomedical research. There are many different laboratory strains of rats and mice, and although it may be unlikely that different strains will exhibit significant transporter gene expression differences that affect the pharmacokinetics of most drugs, caution should still be noted. It may be an important factor because differences in gene expression and function (e.g., efflux transporters, cytochrome P450s, and macrophage antibodies) between rodent strains have been identified (Turk et al., 2004; Lash et al., 2014; Jin et al., 2017; Li et al., 2019; Schaffenrath et al., 2021). Unlike in vitro models, there are many more parameters to consider when using in vivo models to evaluate drug penetration across a physiologically intact BTB. For example, the expression and localization of certain transporters at the BTB (e.g., MRP4 and BCRP) can generate results that cannot to translate to humans, as described previously (Koraïchi et al., 2013; Klein et al., 2014; Hau et al., 2021, 2023). Due to differences in MRP4 localization at the BTB between rats, mice, and humans, a promising drug candidate that is a substrate for MRP4 can generate contradictory results in a rodent disposition study. Moreover, there is no known rodent ortholog for some transporters, such as human OAT4, which limits the use of rodent models when studying this transporter or others without a relevant ortholog. In addition, differences in transporter ortholog selectivity and kinetics can affect how well the testicular disposition data correlates between rodents and humans or nonhuman primates. Therefore, it is critical to determine the expression and localization of rodent BTB transporters and functionally characterize them to compare with data from human tissues.

A previous study showed that ENT1 and ENT2 are localized to the basal and apicolateral membranes of SCs in rodents and humans, respectively (Klein et al., 2013). These transporters were shown to be functional in rodent and human SCs (Kato et al., 2005, 2009; Klein et al., 2013; Hau et al., 2020), which led to a recent study that assessed ENT-mediated testicular disposition of clofarabine in Sprague-Dawley rats (Miller et al., 2021a). Clofarabine is a reported ENT substrate (King et al., 2006; Miller et al., 2021b; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/21-673_Clolar_biopharmr.pdf), whereas lamivudine is not (Miller et al., 2020, 2021b), which is why they were chosen as probe compounds by Miller et al. In that study, clofarabine was further confirmed to be a substrate of ENT1 and could be inhibited by an ENT-specific inhibitor, NBMPR, in primary rat SCs and hT-SerCs (Miller et al., 2021a). Rats were dosed with 10 mg/kg NBMPR-P, the phosphorylated prodrug of NBMPR, or vehicle 20 minutes before 10 mg/kg clofarabine, 10 mg/kg lamivudine, or vehicle dosing. These clofarabine and lamivudine doses were sufficient to detect within the testes and were well within the dose ranges of previously published studies (Bonate et al., 2005; Arthaud and Seals, 2007; Li et al., 2020; https://www.accessdata.fda.gov/drugsatfda_docs/nda/2004/21-673_Clolar_biopharmr.pdf; https://www.accessdata.fda.gov/drugsatfda_docs/label/2004/021673lbl.pdf; https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/206510lbl.pdf). Testicular accumulation of clofarabine in Sprague-Dawley rats in the presence of NBMPR compared with the vehicle control, only approached statistical significance. On the other hand, lamivudine was not a substrate of ENT1 or ENT2 and testicular disposition was completely unaffected by NBMPR. Interestingly, clofarabine causes testicular toxicity in rats, mice, and canines even at a fraction of the clinically recommended dose, according to the FDA package insert (https://www.accessdata.fda.gov/drugsatfda_docs/label/2004/021673lbl.pdf). Despite the lack of statistical significance in the study by Miller et al., these cumulative observations suggest that ENT-mediated disposition across the BTB can be an important physiologic and pharmacological pathway for some nucleosides and nucleoside analogs in rodents and humans (Miller et al., 2021a). However, further studies are necessary to determine if ENTs (and other transporters) mediate the transport of other clinically relevant xenobiotics across the in vivo BTB.

Many other xenobiotics are known to induce a pharmacological or toxicological response in the testes of rodents, including adjudin (Su et al., 2011), cisplatin (Vawda, 1994; Sherif et al., 2014; Soni et al., 2016), cyclophosphamide (Trasler et al., 1988; Matsui et al., 1995), doxorubicin (Imahie et al., 1995), (H2-)gamendazole (Tash et al., 2008a,b; Gupta et al., 2011), imatinib (https://www.accessdata.fda.gov/drugsatfda_docs/nda/2001/21-335_Gleevec_pharmr_P1.pdf), and methotrexate (Padmanabhan et al., 2009; Vardi et al., 2009). The major transporters that accept some of these xenobiotics as substrates have been defined using in vitro heterologous expression systems, although the transporters that accept other compounds as substrates, like H2-gamendazole and gamendazole, are unknown. Unfortunately, comparatively few in vitro and in vivo studies have attempted to identify the BTB transport processes for these compounds. Furthermore, there is a critical lack of information on the expression and localization of transporters at the rodent BTB, which is an issue when studying drug disposition across this barrier. Due to the complexity of in vivo pharmacokinetics, it is prudent to first identify major BTB drug transporters with immunolocalization studies and in vitro models then confirm those findings in vivo like with ENT1 and ENT2.

Although transport data collected from in vivo models can closely correlate with clinical observations, conclusions concerning the role(s) of individual transporters require an extensive array of in vitro and in vivo studies. Since most compounds interact with more than one transporter, in vitro transport studies can inform in vivo experiments, and vice versa, to improve model correlation. The most important consideration is that caution must be taken when using rodents to study drug disposition and toxicity in the testes due to the issues outlined here. Another recent concern for in vivo studies is the push by the FDA to reduce, refine, and replace animals in research, meaning alternative methods such as in vitro and in silico modeling to predict the absorption, distribution, metabolism, and excretion (ADME) characteristics of drugs are becoming increasingly popular.

Nonhuman Primate and Other Mammalian Models

Direct drug disposition studies with human patients are limited to highly regulated clinical studies; however, nonhuman primate models are the closest approximation to human genetics and physiology. Therefore, it is expected that most, if not all, BTB transporters will be expressed in the same locations in humans and nonhuman primates. One study found that MRP1 (basal membrane of SCs), MRP4 (basal membrane of SCs), and MRP8 (round spermatids) are expressed in the same location in humans and rhesus macaques (Klein et al., 2014); however, the localization of many other transporters remains undetermined. Like human studies, nonhuman primate studies are also highly regulated, and FDA recommendations to reduce, refine, and replace these animal studies are warranted. Testicular disposition studies to verify transporter function in nonhuman primates would require physical castration or outright sacrifice, which is unsustainable for drug discovery programs that require routine testing. Confirmational testicular disposition studies in nonhuman primate models may be viable for exceptional drug candidates as a penultimate step for an investigational new drug application. Some drugs, such as H2-gamendazole, have been shown to be effective as a contraceptive agent in nonhuman primates, which confirm observations in other species including mice, rats, and rabbits (Gupta et al., 2011). These findings suggest that BTB pharmacokinetics and pharmacodynamics of some xenobiotics can be adequately modeled in different species, which can then be used to develop in silico models for predictive drug discovery, although an extensive array of data is necessary to achieve accurate and precise predictions for different drug scaffolds.

Like other mammalian in vitro models, alternative species such as hamsters, canines, and rabbits have been used to assess testicular toxicity (Heywood and James, 1978; Rao et al., 1982; Morton, 1988; McEuen and Miller, 1991; Foote and Carney, 2000; Shimomura et al., 2004; Noritake et al., 2011; Vidal and Whitney, 2014). However, there is limited information on transporter expression, localization, and selectivity in these species and how their BTB physiology compares relative to humans, nonhuman primates, and rodents. More recently, pharmaceutical companies have begun exploring the idea of performing ADME studies directly in humans using a microtracer approach by dosing patients with subpharmacologically active 14C-labeled drugs and assessing their pharmacokinetic profiles (Young et al., 2023). The human ADME microtracer study is a powerful strategy; however, it does not help in identifying tissue concentrations, which still require mass balance studies in other species. Therefore, rodent, nonhuman primate, and other mammalian models remain important tools to study drug disposition across the BTB, and further investigation into the expression, localization, and function of transporters in these species is warranted.

Conclusions

The BTB limits the penetration and accumulation of many endogenous and exogenous compounds in the testes due to the presence of a physical membrane barrier and the expression and function of various transporters at the basal membrane of SCs. The expression, selectivity, and common localization of BTB uptake transporters, including the OATs, OCTs, OCTNs, and OATPs, is an indirect indicator for testicular drug disposition. Additionally, more promiscuous efflux transporters such as P-gp, BCRP, and the MRPs actively reduce compound accumulation in and through SCs. Several in vitro and in vivo models are available to study the collective role of these transporters as outlined here. However, there are advantages and disadvantages that must be considered with each model as discussed in this review. Multiple models to assess drug transport across the BTB must be used to account for the biologic complexity of this physiologic barrier. For example, assessing drug transport of a test compound in human, nonhuman primate, and rodent SCs can be supplemented with in vivo rodent studies to generate a holistic picture of BTB disposition.

One potential workflow is to first identify if the uptake of a compound into cultured rodent SCs is transporter-mediated, which can then inform a rodent in vivo disposition study like in Miller et al. (Miller et al., 2021a). An in vitro transport study with human SCs can also be performed at the same time to correlate the findings with rodent SC studies. Potential species differences in transporter expression and localization will also be very important to consider at this stage, because transporter-mediated uptake by transporters expressed at the basal or apicolateral membranes when SCs are plated as flat monolayers are indistinguishable. If possible, transport studies with nonhuman primate SCs may also provide additional data to support future disposition studies in nonhuman primates or humans. Another approach is to identify compounds that exert an in vivo pharmacological or toxicological effect in the testes, then assess SC uptake in various in vitro models. This would be an informed approach, in which some compounds may have well-defined transporter pathways that could help identify the transporters expressed in SCs. The latter approach may be preferable until a quality data set is established. In summary, comprehensive preclinical drug transport studies with multiple in vitro and in vivo BTB models are more likely to accurately reflect and predict observations in clinical studies and should become standard practice.

Data Availability

There are no data sets contained within this paper.

Abbreviations

ADME

absorption, distribution, metabolism, and excretion

BCRP

breast cancer resistance protein

BTB

blood-testis barrier

ENT

equilibrative nucleoside transporter

FDA

Food and Drug Administration

hT-SerC

human TERT-immortalized Sertoli cell

MRP

multidrug resistance-associated protein

OAT

organic anion transporter

OATP

organic anion transporting polypeptide

OCT

organic cation transporter

OCTN

organic cation transporter, novel

P-gp

P-glycoprotein

SC

Sertoli cell

TEER

transepithelial electrical resistance

Authorship Contributions

Wrote or contributed to the writing of the manuscript: Hau, Wright, Cherrington.

Footnotes

This work was supported by funding from National Institutes of Health National Institute of General Medical Sciences [Grants R01GM123643 and R01GM129777] and National Institute of Environmental Health Sciences [Grant T32ES007091].

The authors declare no conflicts of interest.

References

  1. Abu Elhija M, Lunenfeld E, Schlatt S, Huleihel M (2012) Differentiation of murine male germ cells to spermatozoa in a soft agar culture system. Asian J Androl 14:285–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. AbuMadighem A, Shuchat S, Lunenfeld E, Yossifon G, Huleihel M (2022) Testis on a chip-a microfluidic three-dimensional culture system for the development of spermatogenesis in-vitro. Biofabrication 14:035044. [DOI] [PubMed] [Google Scholar]
  3. AbuMadighem A, Solomon R, Stepanovsky A, Kapelushnik J, Shi Q, Meese E, Lunenfeld E, Huleihel M (2018) Development of spermatogenesis in vitro in three-dimensional culture from spermatogonial cells of busulfan-treated immature mice. Int J Mol Sci 19:3804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Achilli TM, McCalla S, Meyer J, Tripathi A, Morgan JR (2014) Multilayer spheroids to quantify drug uptake and diffusion in 3D. Mol Pharm 11:2071–2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Agarwal A, Said TM (2004) Carnitines and male infertility. Reprod Biomed Online 8:376–384. [DOI] [PubMed] [Google Scholar]
  6. Alves-Lopes JP, Söder O, Stukenborg JB (2017) Testicular organoid generation by a novel in vitro three-layer gradient system. Biomaterials 130:76–89. [DOI] [PubMed] [Google Scholar]
  7. Angulo C, Castro MA, Rivas CI, Segretain D, Maldonado R, Yañez AJ, Slebe JC, Vera JC, Concha II (2008) Molecular identification and functional characterization of the vitamin C transporters expressed by Sertoli cells. J Cell Physiol 217:708–716. [DOI] [PubMed] [Google Scholar]
  8. Arkoun B, Dumont L, Milazzo JP, Way A, Bironneau A, Wils J, Macé B, Rives N (2015) Retinol improves in vitro differentiation of pre-pubertal mouse spermatogonial stem cells into sperm during the first wave of spermatogenesis. PLoS One 10:e0116660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Arthaud L, Seals R (2007) Comparison of different intravenous and oral dose schedules on clofarabine-induced hematological toxicity in male rats. Blood 110:4524. [Google Scholar]
  10. Baert Y, De Kock J, Alves-Lopes JP, Söder O, Stukenborg JB, Goossens E (2017) Primary human testicular cells self-organize into organoids with testicular properties. Stem Cell Reports 8:30–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Baert Y, Ruetschle I, Cools W, Oehme A, Lorenz A, Marx U, Goossens E, Maschmeyer I (2020) A multi-organ-chip co-culture of liver and testis equivalents: a first step toward a systemic male reprotoxicity model. Hum Reprod 35:1029–1044. [DOI] [PubMed] [Google Scholar]
  12. Bart J, Hollema H, Groen HJ, de Vries EG, Hendrikse NH, Sleijfer DT, Wegman TD, Vaalburg W, van der Graaf WT (2004) The distribution of drug-efflux pumps, P-gp, BCRP, MRP1 and MRP2, in the normal blood-testis barrier and in primary testicular tumours. Eur J Cancer 40:2064–2070. [DOI] [PubMed] [Google Scholar]
  13. Bell MR (2018) Comparing postnatal development of gonadal hormones and associated social behaviors in rats, mice, and humans. Endocrinology 159:2596–2613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Berensztein EB, Belgorosky A, Rivarola MA (1992) Primary culture of prepubertal human testicular cells isolated from testes collected at necropsy. Acta Endocrinol (Copenh) 127:66–71. [DOI] [PubMed] [Google Scholar]
  15. Bergmann M, Dierichs R (1983) Postnatal formation of the blood-testis barrier in the rat with special reference to the initiation of meiosis. Anat Embryol (Berl) 168:269–275. [DOI] [PubMed] [Google Scholar]
  16. Bernard L, Martinat N, Lécureuil C, Crépieux P, Reiter E, Tilloy-Ellul A, Chevalier S, Guillou F (2007) Dichlorodiphenyltrichloroethane impairs follicle-stimulating hormone receptor-mediated signaling in rat Sertoli cells. Reprod Toxicol 23:158–164. [DOI] [PubMed] [Google Scholar]
  17. Bleasby KCastle JCRoberts CJCheng CBailey WJSina JFKulkarni AVHafey MJEvers RJohnson JM, et al. (2006) Expression profiles of 50 xenobiotic transporter genes in humans and pre-clinical species: a resource for investigations into drug disposition. Xenobiotica 36:963–988. [DOI] [PubMed] [Google Scholar]
  18. Boekelheide K, Lee JW, Hall SJ, Rhind NR, Zaret KS (1993) A tumorigenic murine Sertoli cell line that is temperature-sensitive for differentiation. Am J Pathol 143:1159–1168. [PMC free article] [PubMed] [Google Scholar]
  19. Bonate PL, Arthaud L, Stuhler J, Yerino P, Press RJ, Rose JQ (2005) The distribution, metabolism, and elimination of clofarabine in rats. Drug Metab Dispos 33:739–748. [DOI] [PubMed] [Google Scholar]
  20. Bourdon V, Lablack A, Abbe P, Segretain D, Pointis G (1998) Characterization of a clonal Sertoli cell line using adult PyLT transgenic mice. Biol Reprod 58:591–599. [DOI] [PubMed] [Google Scholar]
  21. Brouwer KL, Aleksunes LM, Brandys B, Giacoia GP, Knipp G, Lukacova V, Meibohm B, Nigam SK, Rieder M, de Wildt SN; Pediatric Transporter Working Group (2015) Human ontogeny of drug transporters: review and recommendations of the Pediatric Transporter Working Group. Clin Pharmacol Ther 98:266–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Buch JP, Lamb DJ, Lipshultz LI, Smith RG (1988) Partial characterization of a unique growth factor secreted by human Sertoli cells. Fertil Steril 49:658–665. [PubMed] [Google Scholar]
  23. Byrn RA, Kiessling AA (1998) Analysis of human immunodeficiency virus in semen: indications of a genetically distinct virus reservoir. J Reprod Immunol 41:161–176. [DOI] [PubMed] [Google Scholar]
  24. Cao X, Surma MA, Simons K (2012) Polarized sorting and trafficking in epithelial cells. Cell Res 22:793–805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Carette D, Weider K, Gilleron J, Giese S, Dompierre J, Bergmann M, Brehm R, Denizot JP, Segretain D, Pointis G (2010) Major involvement of connexin 43 in seminiferous epithelial junction dynamics and male fertility. Dev Biol 346:54–67. [DOI] [PubMed] [Google Scholar]
  26. Chandley AC, Speed RM, Leitch AR (1996) Different distributions of homologous chromosomes in adult human Sertoli cells and in lymphocytes signify nuclear differentiation. J Cell Sci 109:773–776. [DOI] [PubMed] [Google Scholar]
  27. Chen H, Gao Y, Mruk DD, Xiao X, John CM, Turek PJ, Lui WY, Lee WM, Silvestrini B, Cheng CY (2017) Rescue of PFOS-induced human Sertoli cell injury by overexpressing a p-FAK-Y407E phosphomimetic mutant. Sci Rep 7:15810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chen H, Lui WY, Mruk DD, Xiao X, Ge R, Lian Q, Lee WM, Silvestrini B, Cheng CY (2018) Monitoring the integrity of the blood-testis barrier (BTB): an in vivo assay. Methods Mol Biol 1748:245–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Cheung KWKvan Groen BDSpaans Evan Borselen MDde Bruijn ACJMSimons-Oosterhuis YTibboel DSamsom JNVerdijk RMSmeets B, et al. (2019) A comprehensive analysis of ontogeny of renal drug transporters: mRNA analyses, quantitative proteomics, and localization. Clin Pharmacol Ther 106:1083–1092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chu X, Bleasby K, Evers R (2013) Species differences in drug transporters and implications for translating preclinical findings to humans. Expert Opin Drug Metab Toxicol 9:237–252. [DOI] [PubMed] [Google Scholar]
  31. Chui K, Trivedi A, Cheng CY, Cherbavaz DB, Dazin PF, Huynh AL, Mitchell JB, Rabinovich GA, Noble-Haeusslein LJ, John CM (2011) Characterization and functionality of proliferative human Sertoli cells. Cell Transplant 20:619–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Contreras-Baeza YSandoval PYAlarcón RGalaz ACortés-Molina FAlegría KBaeza-Lehnert FArce-Molina RGuequén AFlores CA, et al. (2019) Monocarboxylate transporter 4 (MCT4) is a high affinity transporter capable of exporting lactate in high-lactate microenvironments. J Biol Chem 294:20135–20147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Coombs RJ, Jenkins N (1988) Characterization of a plasminogen activator inhibitor induced by glucocorticoids in immature bovine Sertoli cell-enriched cultures. J Endocrinol 117:69–74. [DOI] [PubMed] [Google Scholar]
  34. Cowan G, Childs AJ, Anderson RA, Saunders PT (2010) Establishment of long-term monolayer cultures of somatic cells from human fetal testes and expansion of peritubular myoid cells in the presence of androgen. Reproduction 139:749–757. [DOI] [PubMed] [Google Scholar]
  35. Cudicini C, Lejeune H, Gomez E, Bosmans E, Ballet F, Saez J, Jégou B (1997) Human Leydig cells and Sertoli cells are producers of interleukins-1 and -6. J Clin Endocrinol Metab 82:1426–1433. [DOI] [PubMed] [Google Scholar]
  36. Davidson AG, Bell RJ, Lees GE, Murphy KE (2007) Isolation, culture, and characterization of canine Sertoli cells. In Vitro Cell Dev Biol Anim 43:324–327. [DOI] [PubMed] [Google Scholar]
  37. De Martino C, Marcante ML, Malorni W, Citro G, Floridi A, Barcellona PS, Capanna E (1985) Morphological and karyological studies on in vitro Sertoli cells of adult crab-eating macaques (Macaca fascicularis). Am J Primatol 9:173–187. [DOI] [PubMed] [Google Scholar]
  38. de Michele F, Poels J, Weerens L, Petit C, Evrard Z, Ambroise J, Gruson D, Wyns C (2017) Preserved seminiferous tubule integrity with spermatogonial survival and induction of Sertoli and Leydig cell maturation after long-term organotypic culture of prepubertal human testicular tissue. Hum Reprod 32:32–45. [DOI] [PubMed] [Google Scholar]
  39. Deen GFBroutet NXu WKnust BSesay FRMcDonald SLRErvin EMarrinan JEGaillard PHabib N, et al. (2017) Ebola RNA persistence in semen of Ebola virus disease survivors—final report. N Engl J Med 377:1428–1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Djakiew D, Dym M (1988) Pachytene spermatocyte proteins influence Sertoli cell function. Biol Reprod 39:1193–1205. [DOI] [PubMed] [Google Scholar]
  41. Dumont L, Arkoun B, Jumeau F, Milazzo JP, Bironneau A, Liot D, Wils J, Rondanino C, Rives N (2015) Assessment of the optimal vitrification protocol for pre-pubertal mice testes leading to successful in vitro production of flagellated spermatozoa. Andrology 3:611–625. [DOI] [PubMed] [Google Scholar]
  42. Dumont L, Oblette A, Rondanino C, Jumeau F, Bironneau A, Liot D, Duchesne V, Wils J, Rives N (2016) Vitamin A prevents round spermatid nuclear damage and promotes the production of motile sperm during in vitro maturation of vitrified pre-pubertal mouse testicular tissue. Mol Hum Reprod 22:819–832. [DOI] [PubMed] [Google Scholar]
  43. Dutertre M, Rey R, Porteu A, Josso N, Picard JY (1997) A mouse Sertoli cell line expressing anti-Müllerian hormone and its type II receptor. Mol Cell Endocrinol 136:57–65. [DOI] [PubMed] [Google Scholar]
  44. Dym M, Fawcett DW (1970) The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium. Biol Reprod 3:308–326. [DOI] [PubMed] [Google Scholar]
  45. Edmonds ME, Woodruff TK (2020) Testicular organoid formation is a property of immature somatic cells, which self-assemble and exhibit long-term hormone-responsive endocrine function. Biofabrication 12:045002. [DOI] [PubMed] [Google Scholar]
  46. Enders GC, Henson JH, Millette CF (1986) Sertoli cell binding to isolated testicular basement membrane. J Cell Biol 103:1109–1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Enokizono J, Kusuhara H, Sugiyama Y (2007) Effect of breast cancer resistance protein (Bcrp/Abcg2) on the disposition of phytoestrogens. Mol Pharmacol 72:967–975. [DOI] [PubMed] [Google Scholar]
  48. Evers R, Piquette-Miller M, Polli JW, Russel FGM, Sprowl JA, Tohyama K, Ware JA, de Wildt SN, Xie W, Brouwer KL; Rand International Transporter Consortium (2018) Disease-associated changes in drug transporters may impact the pharmacokinetics and/or toxicity of drugs: a white paper from the International Transporter Consortium. Clin Pharmacol Ther 104:900–915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Felmlee MA, Zhang Q (2022) Ontogeny of drug transporters, in Drug Transporters (You G and Morris ME eds) pp 393–412, John Wiley & Sons, Inc., New Jersey. [Google Scholar]
  50. Fietz DBakhaus KWapelhorst BGrosser GGünther SAlber JDöring BKliesch SWeidner WGaluska CE, et al. (2013) Membrane transporters for sulfated steroids in the human testis--cellular localization, expression pattern and functional analysis. PLoS One 8:e62638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Fijak M, Meinhardt A (2006) The testis in immune privilege. Immunol Rev 213:66–81. [DOI] [PubMed] [Google Scholar]
  52. Fiorini C, Gilleron J, Carette D, Valette A, Tilloy A, Chevalier S, Segretain D, Pointis G (2008) Accelerated internalization of junctional membrane proteins (connexin 43, N-cadherin and ZO-1) within endocytic vacuoles: an early event of DDT carcinogenicity. Biochim Biophys Acta 1778:56–67. [DOI] [PubMed] [Google Scholar]
  53. Fiorini C, Tilloy-Ellul A, Chevalier S, Charuel C, Pointis G (2004) Sertoli cell junctional proteins as early targets for different classes of reproductive toxicants. Reprod Toxicol 18:413–421. [DOI] [PubMed] [Google Scholar]
  54. Fisher D, Mosaval F, Tharp DL, Bowles DK, Henkel R (2019) Oleanolic acid causes reversible contraception in male mice by increasing the permeability of the germinal epithelium. Reprod Fertil Dev 31:1589–1596. [DOI] [PubMed] [Google Scholar]
  55. Floerl S, Kuehne A, Hagos Y (2020) Functional and pharmacological comparison of human, mouse, and rat organic cation transporter 1 toward drug and pesticide interaction. Int J Mol Sci 21:6871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Foote RH, Carney EW (2000) The rabbit as a model for reproductive and developmental toxicity studies. Reprod Toxicol 14:477–493. [DOI] [PubMed] [Google Scholar]
  57. Franks LM (1968) Spontaneous interstitial and Sertoli cell tumors of a testis in a C3H mouse. Cancer Res 28:125–127. [PubMed] [Google Scholar]
  58. Furuya S, Kumamoto Y, Sugiyama S (1978) Fine structure and development of Sertoli junctions in human testis. Arch Androl 1:211–219. [DOI] [PubMed] [Google Scholar]
  59. Gassei K, Ehmcke J, Schlatt S (2008) Initiation of testicular tubulogenesis is controlled by neurotrophic tyrosine receptor kinases in a three-dimensional Sertoli cell aggregation assay. Reproduction 136:459–469. [DOI] [PubMed] [Google Scholar]
  60. Gassei K, Ehmcke J, Wood MA, Walker WH, Schlatt S (2010) Immature rat seminiferous tubules reconstructed in vitro express markers of Sertoli cell maturation after xenografting into nude mouse hosts. Mol Hum Reprod 16:97–110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Gaur M, Ramathal C, Reijo Pera RA, Turek PJ, John CM (2018) Isolation of human testicular cells and co-culture with embryonic stem cells. Reproduction 155:153–166. [DOI] [PubMed] [Google Scholar]
  62. Ge X, Pan P, Jing J, Hu X, Chen L, Qiu X, Ma R, Jueraitetibaike K, Huang X, Yao B (2018) Rosiglitazone ameliorates palmitic acid-induced cytotoxicity in TM4 Sertoli cells. Reprod Biol Endocrinol 16:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Gholami K, Pourmand G, Koruji M, Sadighigilani M, Navid S, Izadyar F, Abbasi M (2018) Efficiency of colony formation and differentiation of human spermatogenic cells in two different culture systems. Reprod Biol 18:397–403. [DOI] [PubMed] [Google Scholar]
  64. Gohbara A, Katagiri K, Sato T, Kubota Y, Kagechika H, Araki Y, Araki Y, Ogawa T (2010) In vitro murine spermatogenesis in an organ culture system. Biol Reprod 83:261–267. [DOI] [PubMed] [Google Scholar]
  65. Guibert E, Brière S, Pelletier R, Brillard JP, Froment P (2011) Characterization of chicken Sertoli cells in vitro. Poult Sci 90:1276–1286. [DOI] [PubMed] [Google Scholar]
  66. Guo Y, Hai Y, Yao C, Chen Z, Hou J, Li Z, He Z (2015) Long-term culture and significant expansion of human Sertoli cells whilst maintaining stable global phenotype and AKT and SMAD1/5 activation. Cell Commun Signal 13:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Gupta VG, Zelinski M, Steinmetz K, Miklos A, Broward M, Jakkaraj S, Chakrasali R, Georg GI, Tash JS (2011) Anti-spermatogenic efficacy of nonhormonal male contraceptive agent, H2-gamendazole, in mice, rats, rabbits, and nonhuman primates (rhesus), and a multiple low dose oral regimen that gives 100% infertility with complete reversibility. Biol Reprod 85:5. [Google Scholar]
  68. Hadley MA, Byers SW, Suárez-Quian CA, Kleinman HK, Dym M (1985) Extracellular matrix regulates Sertoli cell differentiation, testicular cord formation, and germ cell development in vitro. J Cell Biol 101:1511–1522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Hadley MA, Weeks BS, Kleinman HK, Dym M (1990) Laminin promotes formation of cord-like structures by Sertoli cells in vitro. Dev Biol 140:318–327. [DOI] [PubMed] [Google Scholar]
  70. Harris S, Hermsen SA, Yu X, Hong SW, Faustman EM (2015) Comparison of toxicogenomic responses to phthalate ester exposure in an organotypic testis co-culture model and responses observed in vivo. Reprod Toxicol 58:149–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Harris S, Shubin SP, Wegner S, Van Ness K, Green F, Hong SW, Faustman EM (2016) The presence of macrophages and inflammatory responses in an in vitro testicular co-culture model of male reproductive development enhance relevance to in vivo conditions. Toxicol In Vitro 36:210–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hau RK, Klein RR, Wright SH, Cherrington NJ (2022a) Localization of xenobiotic transporters expressed at the human blood-testis barrier. Drug Metab Dispos 50:770–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Hau RK, Miller SR, Cherrington NJ (2021) Implications of species differences in function and localization of transporters at the blood-testis barrier. Toxicol Sci 181:1–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Hau RK, Miller SR, Wright SH, Cherrington NJ (2020) Generation of a hTERT-immortalized human sertoli cell model to study transporter dynamics at the blood-testis barrier. Pharmaceutics 12:1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Hau RK, Tash JS, Georg GI, Wright SH, Cherrington NJ (2022b) Physiological characterization of the transporter-mediated uptake of the reversible male contraceptive H2-gamendazole across the blood-testis barrier. J Pharmacol Exp Ther 382:299–312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Hau RK, Wright SH, Cherrington NJ (2023) Drug transporters at the human blood-testis barrier. Drug Metab Dispos 51:560–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hayflick L (1965) The limited in vitro lifetime of human diploid cell strains. Exp Cell Res 37:614–636. [DOI] [PubMed] [Google Scholar]
  78. He Z, Kokkinaki M, Jiang J, Dobrinski I, Dym M (2010) Isolation, characterization, and culture of human spermatogonia. Biol Reprod 82:363–372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Heywood R, James RW (1978) Assessment of testicular toxicity in laboratory animals. Environ Health Perspect 24:73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Hilgendorf C, Spahn-Langguth H, Regårdh CG, Lipka E, Amidon GL, Langguth P (2000) Caco-2 versus Caco-2/HT29-MTX co-cultured cell lines: permeabilities via diffusion, inside- and outside-directed carrier-mediated transport. J Pharm Sci 89:63–75. [DOI] [PubMed] [Google Scholar]
  81. Hofmann MC, Van Der Wee KS, Dargart JL, Dirami G, Dettin L, Dym M (2003) Establishment and characterization of neonatal mouse sertoli cell lines. J Androl 24:120–130. [PubMed] [Google Scholar]
  82. Hogarth C (2015) 9 - Retinoic acid metabolism, signaling, and function in the adult testis, in Sertoli Cell Biology, 2nd ed (Griswold MD, ed) pp 247–272, Academic Press, Oxford. [Google Scholar]
  83. Holmes SD, Lipshultz LI, Smith RG (1983) Effect of cannabinoids on human Sertoli cell function in vitro. Arch Androl 11:245–251. [DOI] [PubMed] [Google Scholar]
  84. Hoque MT, Kis O, De Rosa MF, Bendayan R (2015) Raltegravir permeability across blood-tissue barriers and the potential role of drug efflux transporters. Antimicrob Agents Chemother 59:2572–2582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Houzet L, Matusali G, Dejucq-Rainsford N (2014) Origins of HIV-infected leukocytes and virions in semen. J Infect Dis 210(Suppl 3):S622–S630. [DOI] [PubMed] [Google Scholar]
  86. Hu S, Zhu L, Song Y, Zhao X, Chen Q, Pan Y, Zhang J, Bai Y, Zhang H, Shao C (2021) Radiation-induced abscopal reproductive effect is driven by TNF-α/p38 MAPK/Rac1 axis in Sertoli cells. Theranostics 11:5742–5758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Hu Y, Wang R, Xiang Z, Qian W, Han X, Li D (2014) Mixture effects of nonylphenol and di-n-butyl phthalate (monobutyl phthalate) on the tight junctions between Sertoli cells in male rats in vitro and in vivo. Exp Toxicol Pathol 66:445–454. [DOI] [PubMed] [Google Scholar]
  88. Huang S, Ye J, Yu J, Chen L, Zhou L, Wang H, Li Z, Wang C (2014) The accumulation and efflux of lead partly depend on ATP-dependent efflux pump-multidrug resistance protein 1 and glutathione in testis Sertoli cells. Toxicol Lett 226:277–284. [DOI] [PubMed] [Google Scholar]
  89. Huang YHoque MTJenabian MAVyboh KWhyte SKSheehan NLBrassard PBélanger MChomont NFletcher CV, et al. (2016) Antiretroviral drug transporters and metabolic enzymes in human testicular tissue: potential contribution to HIV-1 sanctuary site. J Antimicrob Chemother 71:1954–1965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Huber RD, Gao B, Sidler Pfändler MA, Zhang-Fu W, Leuthold S, Hagenbuch B, Folkers G, Meier PJ, Stieger B (2007) Characterization of two splice variants of human organic anion transporting polypeptide 3A1 isolated from human brain. Am J Physiol Cell Physiol 292:C795–C806. [DOI] [PubMed] [Google Scholar]
  91. Imahie H, Adachi T, Nakagawa Y, Nagasaki T, Yamamura T, Hori M (1995) Effects of adriamycin, an anticancer drug showing testicular toxicity, on fertility in male rats. J Toxicol Sci 20:183–193. [DOI] [PubMed] [Google Scholar]
  92. Jabari ASadighi Gilani MAKoruji MGholami KMohsenzadeh MRastegar TKhadivi FGhanami Gashti NNikmahzar AMojaverrostami S, et al. (2020) Three-dimensional co-culture of human spermatogonial stem cells with Sertoli cells in soft agar culture system supplemented by growth factors and Laminin. Acta Histochem 122:151572. [DOI] [PubMed] [Google Scholar]
  93. Janecki A, Jakubowiak A, Steinberger A (1987) Study of the dynamics of Sertoli cell secretions in a new superfusion, two-compartment culture system. In Vitro Cell Dev Biol 23:492–500. [DOI] [PubMed] [Google Scholar]
  94. Janecki A, Jakubowiak A, Steinberger A (1991a) Effects of cyclic AMP and phorbol ester on transepithelial electrical resistance of Sertoli cell monolayers in two-compartment culture. Mol Cell Endocrinol 82:61–69. [DOI] [PubMed] [Google Scholar]
  95. Janecki A, Jakubowiak A, Steinberger A (1991b) Regulation of transepithelial electrical resistance in two-compartment Sertoli cell cultures: in vitro model of the blood-testis barrier. Endocrinology 129:1489–1496. [DOI] [PubMed] [Google Scholar]
  96. Janecki A, Steinberger A (1986) Polarized Sertoli cell functions in a new two-compartment culture system. J Androl 7:69–71. [DOI] [PubMed] [Google Scholar]
  97. Janecki A, Steinberger A (1987) Bipolar secretion of androgen-binding protein and transferrin by Sertoli cells cultured in a two-compartment culture chamber. Endocrinology 120:291–298. [DOI] [PubMed] [Google Scholar]
  98. Jesus TT, Oliveira PF, Silva J, Barros A, Ferreira R, Sousa M, Cheng CY, Silva BM, Alves MG (2016) Mammalian target of rapamycin controls glucose consumption and redox balance in human Sertoli cells. Fertil Steril 105:825–833.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Jiang C, Hall SJ, Boekelheide K (1997) Development and characterization of a prepubertal rat Sertoli cell line, 93RS2. J Androl 18:393–399. [PubMed] [Google Scholar]
  100. Jin ZL, Chen XF, Ran YH, Li XR, Xiong J, Zheng YY, Gao NN, Li YF (2017) Mouse strain differences in SSRI sensitivity correlate with serotonin transporter binding and function. Sci Rep 7:8631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Jørgensen A, Nielsen JE, Perlman S, Lundvall L, Mitchell RT, Juul A, Rajpert-De Meyts E (2015) Ex vivo culture of human fetal gonads: manipulation of meiosis signalling by retinoic acid treatment disrupts testis development. Hum Reprod 30:2351–2363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Jørgensen A, Young J, Nielsen JE, Joensen UN, Toft BG, Rajpert-De Meyts E, Loveland KL (2014) Hanging drop cultures of human testis and testis cancer samples: a model used to investigate activin treatment effects in a preserved niche. Br J Cancer 110:2604–2614. [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Kabbesh H, Riaz MA, Jensen AD, Scheiner-Bobis G, Konrad L (2021) Long-term maintenance of viable adult rat sertoli cells able to establish testis barrier components and function in response to androgens. Cells 10:2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Kaitu’u-Lino TJ, Sluka P, Foo CF, Stanton PG (2007) Claudin-11 expression and localisation is regulated by androgens in rat Sertoli cells in vitro. Reproduction 133:1169–1179. [DOI] [PubMed] [Google Scholar]
  105. Karacaoğlu E (2022) Flusilazole-induced damage to SerW3 cells via cytotoxicity, oxidative stress and lipid metabolism: An in vitro study. Pestic Biochem Physiol 180:104998. [DOI] [PubMed] [Google Scholar]
  106. Kato R, Maeda T, Akaike T, Tamai I (2005) Nucleoside transport at the blood-testis barrier studied with primary-cultured Sertoli cells. J Pharmacol Exp Ther 312:601–608. [DOI] [PubMed] [Google Scholar]
  107. Kato R, Maeda T, Akaike T, Tamai I (2006) Characterization of novel Na+-dependent nucleobase transport systems at the blood-testis barrier. Am J Physiol Endocrinol Metab 290:E968–E975. [DOI] [PubMed] [Google Scholar]
  108. Kato R, Maeda T, Akaike T, Tamai I (2009) Characterization of nucleobase transport by mouse Sertoli cell line TM4. Biol Pharm Bull 32:450–455. [DOI] [PubMed] [Google Scholar]
  109. Kiessling AA, Fitzgerald LM, Zhang D, Chhay H, Brettler D, Eyre RC, Steinberg J, McGowan K, Byrn RA (1998) Human immunodeficiency virus in semen arises from a genetically distinct virus reservoir. AIDS Res Hum Retroviruses 14(Suppl 1):S33–S41. [PubMed] [Google Scholar]
  110. King KM, Damaraju VL, Vickers MF, Yao SY, Lang T, Tackaberry TE, Mowles DA, Ng AM, Young JD, Cass CE (2006) A comparison of the transportability, and its role in cytotoxicity, of clofarabine, cladribine, and fludarabine by recombinant human nucleoside transporters produced in three model expression systems. Mol Pharmacol 69:346–353. [DOI] [PubMed] [Google Scholar]
  111. Klaassen CD, Aleksunes LM (2010) Xenobiotic, bile acid, and cholesterol transporters: function and regulation. Pharmacol Rev 62:1–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Klein DM, Evans KK, Hardwick RN, Dantzler WH, Wright SH, Cherrington NJ (2013) Basolateral uptake of nucleosides by Sertoli cells is mediated primarily by equilibrative nucleoside transporter 1. J Pharmacol Exp Ther 346:121–129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Klein DM, Wright SH, Cherrington NJ (2014) Localization of multidrug resistance-associated proteins along the blood-testis barrier in rat, macaque, and human testis. Drug Metab Dispos 42:89–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Kobayashi D, Goto A, Maeda T, Nezu J, Tsuji A, Tamai I (2005) OCTN2-mediated transport of carnitine in isolated Sertoli cells. Reproduction 129:729–736. [DOI] [PubMed] [Google Scholar]
  115. Komeya MKimura HNakamura HYokonishi TSato TKojima KHayashi KKatagiri KYamanaka HSanjo H, et al. (2016) Long-term ex vivo maintenance of testis tissues producing fertile sperm in a microfluidic device. Sci Rep 6:21472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Konrad L, Munir Keilani M, Cordes A, Völck-Badouin E, Laible L, Albrecht M, Renneberg H, Aumüller G (2005) Rat Sertoli cells express epithelial but also mesenchymal genes after immortalization with SV40. Biochim Biophys Acta 1722:6–14. [DOI] [PubMed] [Google Scholar]
  117. Koraïchi F, Inoubli L, Lakhdari N, Meunier L, Vega A, Mauduit C, Benahmed M, Prouillac C, Lecoeur S (2013) Neonatal exposure to zearalenone induces long term modulation of ABC transporter expression in testis. Toxicology 310:29–38. [DOI] [PubMed] [Google Scholar]
  118. Kubo Y, Ishizuka S, Ito T, Yoneyama D, Akanuma SI, Hosoya KI (2022) Involvement of TauT/SLC6A6 in taurine transport at the blood-testis barrier. Metabolites 12:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Kullak-Ublick GA, Hagenbuch B, Stieger B, Schteingart CD, Hofmann AF, Wolkoff AW, Meier PJ (1995) Molecular and functional characterization of an organic anion transporting polypeptide cloned from human liver. Gastroenterology 109:1274–1282. [DOI] [PubMed] [Google Scholar]
  120. Lambrot R, Coffigny H, Pairault C, Donnadieu AC, Frydman R, Habert R, Rouiller-Fabre V (2006) Use of organ culture to study the human fetal testis development: effect of retinoic acid. J Clin Endocrinol Metab 91:2696–2703. [DOI] [PubMed] [Google Scholar]
  121. Larsson A, Carlsson L, Gordh T, Lind AL, Thulin M, Kamali-Moghaddam M (2015) The effects of age and gender on plasma levels of 63 cytokines. J Immunol Methods 425:58–61. [DOI] [PubMed] [Google Scholar]
  122. Lash LH, Chiu WA, Guyton KZ, Rusyn I (2014) Trichloroethylene biotransformation and its role in mutagenicity, carcinogenicity and target organ toxicity. Mutat Res Rev Mutat Res 762:22–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Le Magueresse B, Pineau C, Guillou F, Jégou B (1988) Influence of germ cells upon transferrin secretion by rat Sertoli cells in vitro. J Endocrinol 118:R13–R16. [DOI] [PubMed] [Google Scholar]
  124. Lee DR, Kim KS, Yang YH, Oh HS, Lee SH, Chung TG, Cho JH, Kim HJ, Yoon TK, Cha KY (2006a) Isolation of male germ stem cell-like cells from testicular tissue of non-obstructive azoospermic patients and differentiation into haploid male germ cells in vitro. Hum Reprod 21:471–476. [DOI] [PubMed] [Google Scholar]
  125. Lee JH, Gye MC, Choi KW, Hong JY, Lee YB, Park DW, Lee SJ, Min CK (2007) In vitro differentiation of germ cells from nonobstructive azoospermic patients using three-dimensional culture in a collagen gel matrix. Fertil Steril 87:824–833. [DOI] [PubMed] [Google Scholar]
  126. Lee JH, Kim HJ, Kim H, Lee SJ, Gye MC (2006b) In vitro spermatogenesis by three-dimensional culture of rat testicular cells in collagen gel matrix. Biomaterials 27:2845–2853. [DOI] [PubMed] [Google Scholar]
  127. Lee JH, Oh JH, Lee JH, Kim MR, Min CK (2011) Evaluation of in vitro spermatogenesis using poly(D,L-lactic-co-glycolic acid) (PLGA)-based macroporous biodegradable scaffolds. J Tissue Eng Regen Med 5:130–137. [DOI] [PubMed] [Google Scholar]
  128. Legendre A, Froment P, Desmots S, Lecomte A, Habert R, Lemazurier E (2010) An engineered 3D blood-testis barrier model for the assessment of reproductive toxicity potential. Biomaterials 31:4492–4505. [DOI] [PubMed] [Google Scholar]
  129. Li CL, Hsieh CH, Tsai TH (2020) Preclinical pharmacokinetics of lamivudine and its interaction with Schisandra chinensis extract in rats. ACS Omega 5:1997–2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Li F, Ulrich ML, Shih VF, Cochran JH, Hunter JH, Westendorf L, Neale J, Benjamin DR (2019) Mouse strains influence clearance and efficacy of antibody and antibody-drug conjugate via Fc-FcγR interaction. Mol Cancer Ther 18:780–787. [DOI] [PubMed] [Google Scholar]
  131. Li N, Wang T, Han D (2012) Structural, cellular and molecular aspects of immune privilege in the testis. Front Immunol 3:152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Li XWang ZJiang ZGuo JZhang YLi CChung JFolmer JLiu JLian Q, et al. (2016) Regulation of seminiferous tubule-associated stem Leydig cells in adult rat testes. Proc Natl Acad Sci USA 113:2666–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Lipshultz LI, Murthy L, Tindall DJ (1982) Characterization of human Sertoli cells in vitro. J Clin Endocrinol Metab 55:228–237. [DOI] [PubMed] [Google Scholar]
  134. Liu F, Cai C, Wu X, Cheng Y, Lin T, Wei G, He D (2016) Effect of KnockOut serum replacement on germ cell development of immature testis tissue culture. Theriogenology 85:193–199. [DOI] [PubMed] [Google Scholar]
  135. Liu L, Zhu M, Liu X, Fei L, Shen J, Chen D (2021) miR-122-5p regulates the tight junction of the blood-testis barrier of mice via occludin: miR-122-5p can regulate the tight junction. Basic Clin Androl 31:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Liu Y, Fan J, Yan Y, Dang X, Zhao R, Xu Y, Ding Z (2020) JMY expression by Sertoli cells contributes to mediating spermatogenesis in mice. FEBS J 287:5478–5497. [DOI] [PubMed] [Google Scholar]
  137. Ma XGuan CChen RWang YFeng SWang RQu GZhao SWang FWang X, et al. (2021) Pathological and molecular examinations of postmortem testis biopsies reveal SARS-CoV-2 infection in the testis and spermatogenesis damage in COVID-19 patients. Cell Mol Immunol 18:487–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Maeda T, Goto A, Kobayashi D, Tamai I (2007) Transport of organic cations across the blood-testis barrier. Mol Pharm 4:600–607. [DOI] [PubMed] [Google Scholar]
  139. Manning Fox JE, Meredith D, Halestrap AP (2000) Characterisation of human monocarboxylate transporter 4 substantiates its role in lactic acid efflux from skeletal muscle. J Physiol 529:285–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Mao B, Li C, Wen Z, Li H, Wang Y, Chen L, Lian Q, Ge RS (2021) Short-term perfluorooctane sulfonate exposure impairs Leydig cell regeneration in the adult rat testis via targeting hedgehog signaling. Ecotoxicol Environ Saf 214:112121. [DOI] [PubMed] [Google Scholar]
  141. Mather JP (1980) Establishment and characterization of two distinct mouse testicular epithelial cell lines. Biol Reprod 23:243–252. [DOI] [PubMed] [Google Scholar]
  142. Mather JP, Zhuang LZ, Perez-Infante V, Phillips DM (1982) Culture of testicular cells in hormone-supplemented serum-free medium. Ann N Y Acad Sci 383:44–68. [DOI] [PubMed] [Google Scholar]
  143. Matsui H, Mitsumori K, Yasuhara K, Onodera H, Shimo T, Takahashi M (1995) Morphological evaluation of cyclophosphamide testicular toxicity in rats using quantitative morphometry of spermatogenic cycle stages. J Toxicol Sci 20:407–414. [DOI] [PubMed] [Google Scholar]
  144. McCabe MJ, Tarulli GA, Meachem SJ, Robertson DM, Smooker PM, Stanton PG (2010) Gonadotropins regulate rat testicular tight junctions in vivo. Endocrinology 151:2911–2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. McEuen SF, Miller MG (1991) Metabolism and pharmacokinetics of 1,3-dinitrobenzene in the rat and the hamster. Drug Metab Dispos 19:661–666. [PubMed] [Google Scholar]
  146. Megard I, Garrigues A, Orlowski S, Jorajuria S, Clayette P, Ezan E, Mabondzo A (2002) A co-culture-based model of human blood-brain barrier: application to active transport of indinavir and in vivo-in vitro correlation. Brain Res 927:153–167. [DOI] [PubMed] [Google Scholar]
  147. Melaine N, Liénard MO, Dorval I, Le Goascogne C, Lejeune H, Jégou B (2002) Multidrug resistance genes and p-glycoprotein in the testis of the rat, mouse, guinea pig, and human. Biol Reprod 67:1699–1707. [DOI] [PubMed] [Google Scholar]
  148. Merhi RA, Guillaud L, Delouis C, Cotinot C (2001) Establishment and characterization of immortalized ovine Sertoli cell lines. In Vitro Cell Dev Biol Anim 37:581–588. [DOI] [PubMed] [Google Scholar]
  149. Merkl M, Ertl R, Handschuh S, Aurich C, Schäfer-Somi S (2016) The cholesterol transporter ABCA1 is expressed in stallion spermatozoa and reproductive tract tissues. Theriogenology 85:1080–1089. [DOI] [PubMed] [Google Scholar]
  150. Miller SR, Hau RK, Jilek JL, Morales MN, Wright SH, Cherrington NJ (2020) Nucleoside reverse transcriptase inhibitor interaction with human equilibrative nucleoside transporters 1 and 2. Drug Metab Dispos 48:603–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Miller SR, Jilek JL, McGrath ME, Hau RK, Jennings EQ, Galligan JJ, Wright SH, Cherrington NJ (2021a) Testicular disposition of clofarabine in rats is dependent on equilibrative nucleoside transporters. Pharmacol Res Perspect 9:e00831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Miller SRZhang XHau RKJilek JLJennings EQGalligan JJFoil DHZorn KMEkins SWright SH, et al. (2021b) Predicting drug interactions with human equilibrative nucleoside transporters 1 and 2 using functional knockout cell lines and Bayesian modeling. Mol Pharmacol 99:147–162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  153. Mincheva M, Sandhowe-Klaverkamp R, Wistuba J, Redmann K, Stukenborg JB, Kliesch S, Schlatt S (2018) Reassembly of adult human testicular cells: can testis cord-like structures be created in vitro? Mol Hum Reprod 24:55–63. [DOI] [PubMed] [Google Scholar]
  154. Mital P, Hinton BT, Dufour JM (2011) The blood-testis and blood-epididymis barriers are more than just their tight junctions. Biol Reprod 84:851–858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Monet-Kuntz C, Fontaine I (1989) Age-dependent changes in the in-vitro response of a pig Sertoli cell-enriched population to FSH. J Reprod Fertil 86:643–651. [DOI] [PubMed] [Google Scholar]
  156. Mongioi L, Calogero AE, Vicari E, Condorelli RA, Russo GI, Privitera S, Morgia G, La Vignera S (2016) The role of carnitine in male infertility. Andrology 4:800–807. [DOI] [PubMed] [Google Scholar]
  157. Morgan JACheepala SBWang YNeale GAdachi MNachagari DLeggas MZhao WBoyd KVenkataramanan R, et al. (2012) Deregulated hepatic metabolism exacerbates impaired testosterone production in Mrp4-deficient mice. J Biol Chem 287:14456–14466. [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Morgan JALynch JPanetta JCWang YFrase SBao JZheng JOpferman JTJanke LGreen DM, et al. (2015) Apoptosome activation, an important molecular instigator in 6-mercaptopurine induced Leydig cell death. Sci Rep 5:16488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Moroi S, Saitou M, Fujimoto K, Sakakibara A, Furuse M, Yoshida O, Tsukita S (1998) Occludin is concentrated at tight junctions of mouse/rat but not human/guinea pig Sertoli cells in testes. Am J Physiol 274:C1708–C1717. [DOI] [PubMed] [Google Scholar]
  160. Morton D (1988) The use of rabbits in male reproductive toxicology. Environ Health Perspect 77:5–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  161. Mruk DD, Cheng CY (2011) An in vitro system to study Sertoli cell blood-testis barrier dynamics. Methods Mol Biol 763:237–252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. Mruk DD, Cheng CY (2015) The mammalian blood-testis barrier: its biology and regulation. Endocr Rev 36:564–591. [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Mruk DD, Su L, Cheng CY (2011) Emerging role for drug transporters at the blood-testis barrier. Trends Pharmacol Sci 32:99–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Neaves WB (1973) Permeability of Sertoli cell tight junctions to lanthanum after ligation of ductus deferens and ductuli efferentes. J Cell Biol 59:559–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Nesbit ME Jr, Robison LL, Ortega JA, Sather HN, Donaldson M, Hammond D (1980) Testicular relapse in childhood acute lymphoblastic leukemia: association with pretreatment patient characteristics and treatment. A report for Childrens Cancer Study Group. Cancer 45:2009–2016. [DOI] [PubMed] [Google Scholar]
  166. Nicholls PK, Harrison CA, Gilchrist RB, Farnworth PG, Stanton PG (2009) Growth differentiation factor 9 is a germ cell regulator of Sertoli cell function. Endocrinology 150:2481–2490. [DOI] [PubMed] [Google Scholar]
  167. Noritake K, Suzuki J, Matsuoka T, Makino T, Ohnishi H, Shimomura K, Uenoyama Y, Tsukamura H, Maeda K, Sanbuissho A (2011) Testicular toxicity induced by a triple neurokinin receptor antagonist in male dogs. Reprod Toxicol 31:440–446. [DOI] [PubMed] [Google Scholar]
  168. Oliver E, Alves-Lopes JP, Harteveld F, Mitchell RT, Åkesson E, Söder O, Stukenborg JB (2021) Self-organising human gonads generated by a Matrigel-based gradient system. BMC Biol 19:212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Olovnikov AM (1996) Telomeres, telomerase, and aging: origin of the theory. Exp Gerontol 31:443–448. [DOI] [PubMed] [Google Scholar]
  170. Padmanabhan S, Tripathi DN, Vikram A, Ramarao P, Jena GB (2009) Methotrexate-induced cytotoxicity and genotoxicity in germ cells of mice: intervention of folic and folinic acid. Mutat Res 673:43–52. [DOI] [PubMed] [Google Scholar]
  171. Palmero S, Bottazzi C, Costa M, Leone M, Fugassa E (2000) Metabolic effects of L-carnitine on prepubertal rat Sertoli cells. Horm Metab Res 32:87–90. [DOI] [PubMed] [Google Scholar]
  172. Pan F, Chi L, Schlatt S (2013) Effects of nanostructures and mouse embryonic stem cells on in vitro morphogenesis of rat testicular cords. PLoS One 8:e60054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Papadopoulos D, Dietze R, Shihan M, Kirch U, Scheiner-Bobis G (2016) Dehydroepiandrosterone sulfate stimulates expression of blood-testis-barrier proteins claudin-3 and -5 and tight junction formation via a Gnα11-coupled receptor in Sertoli cells. PLoS One 11:e0150143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Paquis-Flucklinger V, Michiels JF, Vidal F, Alquier C, Pointis G, Bourdon V, Cuzin F, Rassoulzadegan M (1993) Expression in transgenic mice of the large T antigen of polyomavirus induces Sertoli cell tumours and allows the establishment of differentiated cell lines. Oncogene 8:2087–2094. [PubMed] [Google Scholar]
  175. Pelletier RM (2011) The blood-testis barrier: the junctional permeability, the proteins and the lipids. Prog Histochem Cytochem 46:49–127. [DOI] [PubMed] [Google Scholar]
  176. Pendergraft SS, Sadri-Ardekani H, Atala A, Bishop CE (2017) Three-dimensional testicular organoid: a novel tool for the study of human spermatogenesis and gonadotoxicity in vitro. Biol Reprod 96:720–732. [DOI] [PubMed] [Google Scholar]
  177. Perrard MH, Sereni N, Schluth-Bolard C, Blondet A, D Estaing SG, Plotton I, Morel-Journel N, Lejeune H, David L, Durand P (2016) Complete human and rat ex vivo spermatogenesis from fresh or frozen testicular tissue. Biol Reprod 95:89. [DOI] [PubMed] [Google Scholar]
  178. Peschon JJ, Behringer RR, Cate RL, Harwood KA, Idzerda RL, Brinster RL, Palmiter RD (1992) Directed expression of an oncogene to Sertoli cells in transgenic mice using mullerian inhibiting substance regulatory sequences. Mol Endocrinol 6:1403–1411. [DOI] [PubMed] [Google Scholar]
  179. Pizzagalli F, Hagenbuch B, Stieger B, Klenk U, Folkers G, Meier PJ (2002) Identification of a novel human organic anion transporting polypeptide as a high affinity thyroxine transporter. Mol Endocrinol 16:2283–2296. [DOI] [PubMed] [Google Scholar]
  180. Pognan F, Masson MT, Lagelle F, Charuel C (1997) Establishment of a rat Sertoli cell line that displays the morphological and some of the functional characteristics of the native cell. Cell Biol Toxicol 13:453–463. [DOI] [PubMed] [Google Scholar]
  181. Politch JA, Mayer KH, Welles SL, O’Brien WX, Xu C, Bowman FP, Anderson DJ (2012) Highly active antiretroviral therapy does not completely suppress HIV in semen of sexually active HIV-infected men who have sex with men. AIDS 26:1535–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Potter SJ, DeFalco T (2015) Using ex vivo upright droplet cultures of whole fetal organs to study developmental processes during mouse organogenesis. J Vis Exp 105:e53262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Prozialeck WC, Lamar PC (1997) Cadmium (Cd2+) disrupts E-cadherin-dependent cell-cell junctions in MDCK cells. In Vitro Cell Dev Biol Anim 33:516–526. [DOI] [PubMed] [Google Scholar]
  184. Qian X, Mruk DD, Wong EW, Cheng CY (2013) Breast cancer resistance protein regulates apical ectoplasmic specialization dynamics stage specifically in the rat testis. Am J Physiol Endocrinol Metab 304:E757–E769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Qiu R, Horvath A, Stahlmann R (2016) Effects of four nucleoside analogues used as antiviral agents on rat Sertoli cells (SerW3) in vitro. Arch Toxicol 90:1975–1981. [DOI] [PubMed] [Google Scholar]
  186. Rao KS, Burek JD, Murray FJ, John JA, Schwetz BA, Beyer JE, Parker CM (1982) Toxicologic and reproductive effects of inhaled 1,2-dibromo-3-chloropropane in male rabbits. Fundam Appl Toxicol 2:241–251. [DOI] [PubMed] [Google Scholar]
  187. Reda A, Albalushi H, Montalvo SC, Nurmio M, Sahin Z, Hou M, Geijsen N, Toppari J, Söder O, Stukenborg JB (2017) Knock-out serum replacement and melatonin effects on germ cell differentiation in murine testicular explant cultures. Ann Biomed Eng 45:1783–1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Reda A, Hou M, Landreh L, Kjartansdóttir KR, Svechnikov K, Söder O, Stukenborg JB (2014) In vitro spermatogenesis—optimal culture conditions for testicular cell survival, germ cell differentiation, and steroidogenesis in rats. Front Endocrinol (Lausanne) 5:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Reda A, Hou M, Winton TR, Chapin RE, Söder O, Stukenborg JB (2016) In vitro differentiation of rat spermatogonia into round spermatids in tissue culture. Mol Hum Reprod 22:601–612. [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Renier G, Gaulin J, Gibb W, Simard P, Leung TK, Collu R, Ducharme JR (1986) Isolation, purification and culture of Sertoli cells from immature piglet testes. Acta Endocrinol (Copenh) 111:411–418. [DOI] [PubMed] [Google Scholar]
  191. Reuter K, Ehmcke J, Stukenborg JB, Simoni M, Damm OS, Redmann K, Schlatt S, Wistuba J (2014) Reassembly of somatic cells and testicular organogenesis in vitro. Tissue Cell 46:86–96. [DOI] [PubMed] [Google Scholar]
  192. Richer G, Hobbs RM, Loveland KL, Goossens E, Baert Y (2021) Long-term maintenance and meiotic entry of early germ cells in murine testicular organoids functionalized by 3D printed scaffolds and air-medium interface cultivation. Front Physiol 12:757565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Ritzén EM (1990) Testicular relapse of acute lymphoblastic leukemia (ALL). J Reprod Immunol 18:117–121. [DOI] [PubMed] [Google Scholar]
  194. Roberts KP, Banerjee PP, Tindall JW, Zirkin BR (1995) Immortalization and characterization of a Sertoli cell line from the adult rat. Biol Reprod 53:1446–1453. [DOI] [PubMed] [Google Scholar]
  195. Robillard KR, Hoque T, Bendayan R (2012) Expression of ATP-binding cassette membrane transporters in rodent and human sertoli cells: relevance to the permeability of antiretroviral therapy at the blood-testis barrier. J Pharmacol Exp Ther 340:96–108. [DOI] [PubMed] [Google Scholar]
  196. Robinson CLChong ACNAshbrook AWJeng GJin JChen HTang EIMartin LAKim RSKenyon RM, et al. (2018) Male germ cells support long-term propagation of Zika virus. Nat Commun 9:2090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Rondanino C, Maouche A, Dumont L, Oblette A, Rives N (2017) Establishment, maintenance and functional integrity of the blood-testis barrier in organotypic cultures of fresh and frozen/thawed prepubertal mouse testes. Mol Hum Reprod 23:304–320. [DOI] [PubMed] [Google Scholar]
  198. Roulet V, Denis H, Staub C, Le Tortorec A, Delaleu B, Satie AP, Patard JJ, Jégou B, Dejucq-Rainsford N (2006) Human testis in organotypic culture: application for basic or clinical research. Hum Reprod 21:1564–1575. [DOI] [PubMed] [Google Scholar]
  199. Russell LD, Bartke A, Goh JC (1989) Postnatal development of the Sertoli cell barrier, tubular lumen, and cytoskeleton of Sertoli and myoid cells in the rat, and their relationship to tubular fluid secretion and flow. Am J Anat 184:179–189. [DOI] [PubMed] [Google Scholar]
  200. Sakib S, Uchida A, Valenzuela-Leon P, Yu Y, Valli-Pulaski H, Orwig K, Ungrin M, Dobrinski I (2019) Formation of organotypic testicular organoids in microwell culture. Biol Reprod 100:1648–1660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Santiemma V, Rosati P, Guerzoni C, Mariani S, Beligotti F, Magnanti M, Garufi G, Galoni T, Fabbrini A (1992) Human Sertoli cells in vitro: morphological features and androgen-binding protein secretion. J Steroid Biochem Mol Biol 43:423–429. [DOI] [PubMed] [Google Scholar]
  202. Sasaki S, Futagi Y, Ideno M, Kobayashi M, Narumi K, Furugen A, Iseki K (2016) Effect of diclofenac on SLC16A3/MCT4 by the Caco-2 cell line. Drug Metab Pharmacokinet 31:218–223. [DOI] [PubMed] [Google Scholar]
  203. Sato T, Katagiri K, Gohbara A, Inoue K, Ogonuki N, Ogura A, Kubota Y, Ogawa T (2011a) In vitro production of functional sperm in cultured neonatal mouse testes. Nature 471:504–507. [DOI] [PubMed] [Google Scholar]
  204. Sato T, Katagiri K, Kojima K, Komeya M, Yao M, Ogawa T (2015) In vitro spermatogenesis in explanted adult mouse testis tissues. PLoS One 10:e0130171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  205. Sato T, Katagiri K, Yokonishi T, Kubota Y, Inoue K, Ogonuki N, Matoba S, Ogura A, Ogawa T (2011b) In vitro production of fertile sperm from murine spermatogonial stem cell lines. Nat Commun 2:472. [DOI] [PubMed] [Google Scholar]
  206. Sato Y, Yoshida K, Nozawa S, Yoshiike M, Arai M, Otoi T, Iwamoto T (2013) Establishment of adult mouse Sertoli cell lines by using the starvation method. Reproduction 145:505–516. [DOI] [PubMed] [Google Scholar]
  207. Schaffenrath J, Huang SF, Wyss T, Delorenzi M, Keller A (2021) Characterization of the blood-brain barrier in genetically diverse laboratory mouse strains. Fluids Barriers CNS 18:34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  208. Schrade AKyrönlahti AAkinrinade OPihlajoki MFischer SRodriguez VMOtte KVelagapudi VToppari JWilson DB, et al. (2016) GATA4 regulates blood-testis barrier function and lactate metabolism in mouse Sertoli cells. Endocrinology 157:2416–2431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  209. Seidl K, Holstein AF (1990) Organ culture of human seminiferous tubules: a useful tool to study the role of nerve growth factor in the testis. Cell Tissue Res 261:539–547. [DOI] [PubMed] [Google Scholar]
  210. Selva DM, Hirsch-Reinshagen V, Burgess B, Zhou S, Chan J, McIsaac S, Hayden MR, Hammond GL, Vogl AW, Wellington CL (2004) The ATP-binding cassette transporter 1 mediates lipid efflux from Sertoli cells and influences male fertility. J Lipid Res 45:1040–1050. [DOI] [PubMed] [Google Scholar]
  211. Sherif IO, Abdel-Aziz A, Sarhan OM (2014) Cisplatin-induced testicular toxicity in rats: the protective effect of arjunolic acid. J Biochem Mol Toxicol 28:515–521. [DOI] [PubMed] [Google Scholar]
  212. Shimomura K, Shimada M, Hagiwara M, Harada S, Kato M, Furuhama K (2004) Testicular toxicity induced in dogs by nefiracetam, a neutrotransmission enhancer. Reprod Toxicol 18:423–430. [DOI] [PubMed] [Google Scholar]
  213. Siemann DN, Strange DP, Maharaj PN, Shi PY, Verma S (2017) Zika virus infects human Sertoli cells and modulates the integrity of the in vitro blood-testis barrier model. J Virol 91::e00623-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Skinner MK, Tung PS, Fritz IB (1985) Cooperativity between Sertoli cells and testicular peritubular cells in the production and deposition of extracellular matrix components. J Cell Biol 100:1941–1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  215. Soni KK, Kim HK, Choi BR, Karna KK, You JH, Cha JS, Shin YS, Lee SW, Kim CY, Park JK (2016) Dose-dependent effects of cisplatin on the severity of testicular injury in Sprague Dawley rats: reactive oxygen species and endoplasmic reticulum stress. Drug Des Devel Ther 10:3959–3968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  216. Srinivasan B, Kolli AR, Esch MB, Abaci HE, Shuler ML, Hickman JJ (2015) TEER measurement techniques for in vitro barrier model systems. J Lab Autom 20:107–126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  217. Stallard BJ, Griswold MD (1990) Germ cell regulation of Sertoli cell transferrin mRNA levels. Mol Endocrinol 4:393–401. [DOI] [PubMed] [Google Scholar]
  218. Steinberger A, Klinefelter G (1993) Sensitivity of Sertoli and Leydig cells to xenobiotics in in vitro models. Reprod Toxicol 7 Suppl 1):23–37. [DOI] [PubMed] [Google Scholar]
  219. Steinberger A, Steinberger E (1965) Differentiation of rat seminiferous epithelium in organ culture. J Reprod Fertil 9:243–248. [DOI] [PubMed] [Google Scholar]
  220. Steinberger A, Steinberger E, Perloff WH (1964) Mammalian testes in organ culture. Exp Cell Res 36:19–27. [DOI] [PubMed] [Google Scholar]
  221. Stukenborg JB, Wistuba J, Luetjens CM, Elhija MA, Huleihel M, Lunenfeld E, Gromoll J, Nieschlag E, Schlatt S (2008) Coculture of spermatogonia with somatic cells in a novel three-dimensional soft-agar-culture-system. J Androl 29:312–329. [DOI] [PubMed] [Google Scholar]
  222. Su L, Mruk DD, Cheng CY (2012) Regulation of drug transporters in the testis by environmental toxicant cadmium, steroids and cytokines. Spermatogenesis 2:285–293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  223. Su L, Mruk DD, Lee WM, Cheng CY (2011) Drug transporters and blood—testis barrier function. J Endocrinol 209:337–351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Suzuki S, Sato K (2003) The fertilising ability of spermatogenic cells derived from cultured mouse immature testicular tissue. Zygote 11:307–316. [DOI] [PubMed] [Google Scholar]
  225. Suzuki TOnogawa TAsano NMizutamari HMikkaichi TTanemoto MAbe MSatoh FUnno MNunoki K, et al. (2003) Identification and characterization of novel rat and human gonad-specific organic anion transporters. Mol Endocrinol 17:1203–1215. [DOI] [PubMed] [Google Scholar]
  226. Syvänen S, Lindhe O, Palner M, Kornum BR, Rahman O, Långström B, Knudsen GM, Hammarlund-Udenaes M (2009) Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos 37:635–643. [DOI] [PubMed] [Google Scholar]
  227. Tabuchi Y, Ohta S, Yanai N, Obinata M, Kondo T, Fuse H, Asano S (2002) Development of the conditionally immortalized testicular Sertoli cell line TTE3 expressing Sertoli cell specific genes from mice transgenic for temperature sensitive simian virus 40 large T antigen gene. J Urol 167:1538–1545. [PubMed] [Google Scholar]
  228. Tabuchi Y, Takahashi R, Ueda M, Obinata M (2003) Development of a conditionally immortalized testicular Sertoli cell line RTS3-3 from adult transgenic rats harboring temperature-sensitive simian virus 40 large T-antigen gene. Cell Struct Funct 28:87–95. [DOI] [PubMed] [Google Scholar]
  229. Tamai I, Ohashi R, Nezu J, Yabuuchi H, Oku A, Shimane M, Sai Y, Tsuji A (1998) Molecular and functional identification of sodium ion-dependent, high affinity human carnitine transporter OCTN2. J Biol Chem 273:20378–20382. [DOI] [PubMed] [Google Scholar]
  230. Tarulli GA, Meachem SJ, Schlatt S, Stanton PG (2008) Regulation of testicular tight junctions by gonadotrophins in the adult Djungarian hamster in vivo. Reproduction 135:867–877. [DOI] [PubMed] [Google Scholar]
  231. Tash JS, Attardi B, Hild SA, Chakrasali R, Jakkaraj SR, Georg GI (2008a) A novel potent indazole carboxylic acid derivative blocks spermatogenesis and is contraceptive in rats after a single oral dose. Biol Reprod 78:1127–1138. [DOI] [PubMed] [Google Scholar]
  232. Tash JSChakrasali RJakkaraj SRHughes JSmith SKHornbaker KHeckert LLOzturk SBHadden MKKinzy TG, et al. (2008b) Gamendazole, an orally active indazole carboxylic acid male contraceptive agent, targets HSP90AB1 (HSP90BETA) and EEF1A1 (eEF1A), and stimulates Il1a transcription in rat Sertoli cells. Biol Reprod 78:1139–1152. [DOI] [PubMed] [Google Scholar]
  233. Ter Horst RJaeger MSmeekens SPOosting MSwertz MALi YKumar VDiavatopoulos DAJansen AFMLemmers H, et al. (2016) Host and environmental factors influencing individual human cytokine responses. Cell 167:1111–1124.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  234. Toyama Y, Ohkawa M, Oku R, Maekawa M, Yuasa S (2001) Neonatally administered diethylstilbestrol retards the development of the blood-testis barrier in the rat. J Androl 22:413–423. [PubMed] [Google Scholar]
  235. Trasler JM, Hermo L, Robaire B (1988) Morphological changes in the testis and epididymis of rats treated with cyclophosphamide: a quantitative approach. Biol Reprod 38:463–479. [DOI] [PubMed] [Google Scholar]
  236. Tremblay AR, Delbes G (2018) In vitro study of doxorubicin-induced oxidative stress in spermatogonia and immature Sertoli cells. Toxicol Appl Pharmacol 348:32–42. [DOI] [PubMed] [Google Scholar]
  237. Tung PS, Rosenior J, Fritz IB (1987) Isolation and culture of ram rete testis epithelial cells: structural and biochemical characteristics. Biol Reprod 36:1297–1312. [DOI] [PubMed] [Google Scholar]
  238. Turk R, ’t Hoen PA, Sterrenburg E, de Menezes RX, de Meijer EJ, Boer JM, van Ommen GJ, den Dunnen JT (2004) Gene expression variation between mouse inbred strains. BMC Genomics 5:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. van Groen BDNicolaï JKuik ACVan Cruchten Svan Peer ESmits ASchmidt Sde Wildt SNAllegaert KDe Schaepdrijver L, et al. (2021) Ontogeny of hepatic transporters and drug-metabolizing enzymes in humans and in nonclinical species. Pharmacol Rev 73:597–678. [DOI] [PubMed] [Google Scholar]
  240. Vardi N, Parlakpinar H, Ates B, Cetin A, Otlu A (2009) Antiapoptotic and antioxidant effects of beta-carotene against methotrexate-induced testicular injury. Fertil Steril 92:2028–2033. [DOI] [PubMed] [Google Scholar]
  241. Vawda AI (1994) Effect of testosterone on cisplatin-induced testicular damage. Arch Androl 32:53–57. [DOI] [PubMed] [Google Scholar]
  242. Vazeille MC, Chevalier M (1980) [Sertoli cells of the impubescent pig: a method for obtaining isolated cells]. C R Seances Soc Biol Fil 173:1064–1069. [PubMed] [Google Scholar]
  243. Vermeulen M, Del Vento F, de Michele F, Poels J, Wyns C (2018) Development of a cytocompatible scaffold from pig immature testicular tissue allowing human Sertoli cell attachment, proliferation and functionality. Int J Mol Sci 19:227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  244. Vidal JD, Whitney KM (2014) Morphologic manifestations of testicular and epididymal toxicity. Spermatogenesis 4:e979099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  245. von Kopylow K, Schulze W, Salzbrunn A, Schaks M, Schäfer E, Roth B, Schlatt S, Spiess AN (2018) Dynamics, ultrastructure and gene expression of human in vitro organized testis cells from testicular sperm extraction biopsies. Mol Hum Reprod 24:123–134. [DOI] [PubMed] [Google Scholar]
  246. Wang SQian ZGe XLi CXue MLiang KMa ROuyang LZheng LJing J, et al. (2022) LncRNA Tug1 maintains blood-testis barrier integrity by modulating Ccl2 expression in high-fat diet mice. Cell Mol Life Sci 79:114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Wang Y, Ye J, Ganapathy V, Longo N (1999) Mutations in the organic cation/carnitine transporter OCTN2 in primary carnitine deficiency. Proc Natl Acad Sci USA 96:2356–2360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  248. Wen LYuan QSun MNiu MWang HFu HZhou FYao CWang XLi Z, et al. (2017) Generation and characteristics of human Sertoli cell line immortalized by overexpression of human telomerase. Oncotarget 8:16553–16570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  249. Willems A, Batlouni SR, Esnal A, Swinnen JV, Saunders PT, Sharpe RM, França LR, De Gendt K, Verhoeven G (2010) Selective ablation of the androgen receptor in mouse Sertoli cells affects Sertoli cell maturation, barrier formation and cytoskeletal development. PLoS One 5:e14168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  250. Wu D, Huang CJ, Jiao XF, Ding ZM, Zhang JY, Chen F, Wang YS, Li X, Huo LJ (2017a) Olaquindox disrupts tight junction integrity and cytoskeleton architecture in mouse Sertoli cells. Oncotarget 8:88630–88644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  251. Wu D, Huang CJ, Khan FA, Jiao XF, Liu XM, Pandupuspitasari NS, Brohi RD, Huo LJ (2017b) SENP3 grants tight junction integrity and cytoskeleton architecture in mouse Sertoli cells. Oncotarget 8:58430–58442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  252. Xiong Z, Wang C, Wang Z, Dai H, Song Q, Zou Z, Xiao B, Zhao AZ, Bai X, Chen Z (2018) Raptor directs Sertoli cell cytoskeletal organization and polarity in the mouse testis. Biol Reprod 99:1289–1302. [DOI] [PubMed] [Google Scholar]
  253. Yokonishi T, Sato T, Katagiri K, Komeya M, Kubota Y, Ogawa T (2013) In vitro reconstruction of mouse seminiferous tubules supporting germ cell differentiation. Biol Reprod 89:15. [DOI] [PubMed] [Google Scholar]
  254. Yokonishi TSato TKomeya MKatagiri KKubota YNakabayashi KHata KInoue KOgonuki NOgura A, et al. (2014) Offspring production with sperm grown in vitro from cryopreserved testis tissues. Nat Commun 5:4320. [DOI] [PubMed] [Google Scholar]
  255. Young GCSpracklin DKJames ADHvenegaard MGScarfe GWagner DSGeorgi KSchieferstein HBjornsdottir Ivan Groen B, et al. (2023) Considerations for human ADME strategy and design paradigm shift(s)—an industry white paper. Clin Pharmacol Ther 113:775–781. [DOI] [PubMed] [Google Scholar]
  256. Yu J, Sun J, Fan Y, Su J, Xie J, Wu Y, Liu X, Wang C (2019) Exposure to Pb and Cd alters MCT4/CD147 expression and MCT4/CD147-dependent lactate transport in mice Sertoli cells cultured in vitro. Toxicol In Vitro 56:30–40. [DOI] [PubMed] [Google Scholar]
  257. Zenzes MT, Engel W (1981) The capacity of testicular cells of the postnatal rat to reorganize into histotypic structures. Differentiation 20:157–161. [DOI] [PubMed] [Google Scholar]
  258. Zhang H, Liu B, Qiu Y, Fan Jf, Yu Sj (2013) Pure cultures and characterization of yak Sertoli cells. Tissue Cell 45:414–420. [DOI] [PubMed] [Google Scholar]
  259. Zhang J, Hatakeyama J, Eto K, Abe S (2014) Reconstruction of a seminiferous tubule-like structure in a 3 dimensional culture system of re-aggregated mouse neonatal testicular cells within a collagen matrix. Gen Comp Endocrinol 205:121–132. [DOI] [PubMed] [Google Scholar]
  260. Zhang L, Ji X, Ding F, Wu X, Tang N, Wu Q (2022) Apoptosis and blood-testis barrier disruption during male reproductive dysfunction induced by PAHs of different molecular weights. Environ Pollut 300:118959. [DOI] [PubMed] [Google Scholar]
  261. Zhang L, Liang C, Xu P, Liu M, Xu F, Wang X (2019) Characterization of in vitro Mrp2 transporter model based on intestinal organoids. Regul Toxicol Pharmacol 108:104449. [DOI] [PubMed] [Google Scholar]
  262. Zhang X, Wang L, Zhang X, Ren L, Shi W, Tian Y, Zhu J, Zhang T (2017) The use of KnockOut serum replacement (KSR) in three dimensional rat testicular cells co-culture model: An improved male reproductive toxicity testing system. Food Chem Toxicol 106(Pt A):487–495. [DOI] [PubMed] [Google Scholar]
  263. Zhang Z, McGoron AJ, Crumpler ET, Li CZ (2011) Co-culture based blood-brain barrier in vitro model, a tissue engineering approach using immortalized cell lines for drug transport study. Appl Biochem Biotechnol 163:278–295. [DOI] [PubMed] [Google Scholar]
  264. Zhao S, Zhu W, Xue S, Han D (2014) Testicular defense systems: immune privilege and innate immunity. Cell Mol Immunol 11:428–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  265. Zhou Y, Geng X, Chen Y, Shi H, Yang Y, Zhu C, Yu G, Tang Z (2018) Essential roles of Akt/Snail pathway in microcystin-LR-induced tight junction toxicity in Sertoli cell. Food Chem Toxicol 112:290–298. [DOI] [PubMed] [Google Scholar]
  266. Zietek TGiesbertz PEwers MReichart FWeinmüller MUrbauer EHaller DDemir IECeyhan GOKessler H, et al. (2020) Organoids to study intestinal nutrient transport, drug uptake and metabolism—update to the human model and expansion of applications. Front Bioeng Biotechnol 8:577656. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Drug Metabolism and Disposition are provided here courtesy of American Society for Pharmacology and Experimental Therapeutics

RESOURCES