Next Article in Journal
Facile Splint-Free Circularization of ssDNA with T4 DNA Ligase by Redesigning the Linear Substrate to Form an Intramolecular Dynamic Nick
Previous Article in Journal
Facilities in Molecular Biomarkers in Cardiology
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Advancement and Potential Applications of Epididymal Organoids

Institute of Reproductive Medicine, Medical School, Nantong University, Nantong 226019, China
*
Author to whom correspondence should be addressed.
Biomolecules 2024, 14(8), 1026; https://doi.org/10.3390/biom14081026
Submission received: 4 July 2024 / Revised: 4 August 2024 / Accepted: 8 August 2024 / Published: 17 August 2024

Abstract

:
The epididymis, a key reproductive organ, is crucial for sperm concentration, maturation, and storage. Despite a comprehensive understanding of many of its functions, several aspects of the complex processes within the epididymis remain obscure. Dysfunction in this organ is intricately connected to the formation of the microenvironment, disruptions in sperm maturation, and the progression of male infertility. Thus, elucidating the functional mechanisms of the epididymal epithelium is imperative. Given the variety of cell types present within the epididymal epithelium, utilizing a three-dimensional (3D) in vitro model provides a holistic and practical framework for exploring the multifaceted roles of the epididymis. Organoid cell culture, involving the co-cultivation of pluripotent or adult stem cells with growth factors on artificial matrix scaffolds, effectively recreates the in vivo cell growth microenvironment, thereby offering a promising avenue for studying the epididymis. The field of epididymal organoids is relatively new, with few studies focusing on their formation and even fewer detailing the generation of organoids that exhibit epididymis-specific structures and functions. Ongoing challenges in both clinical applications and mechanistic studies underscore the importance of this research. This review summarizes the established methodologies for inducing the in vitro cultivation of epididymal cells, outlines the various approaches for the development of epididymal organoids, and explores their potential applications in the field of male reproductive biology.

1. Introduction

The epididymis is characterized by an intricately coiled and convoluted tubular architecture, primarily segmented into caput, corpus, and cauda [1,2,3,4]. In rodents, an initial segment (IS) is located between the efferent ducts and the caput [5]. The caput, the initial segment, interfaces with the testicular efferent ducts, while the cauda connects to the vas deferens [6]. The length of the epididymis varies among species, exemplified by a human unfolded length of approximately 6 m, where sperm migrate from the caput to the cauda within a period of 1–2 weeks [6,7]. Each of these distinct regions possesses unique anatomical and physiological characteristics. The epithelial cells comprising the epididymal duct wall are its primary cellular component, forming a pseudostratified epithelium composed of multiple cell types (see Figure 1) [8,9,10]. These cell types include principal cells, basal cells, clear cells, apical cells, and narrow cells [6,11]. Principal cells are the predominant type, constituting 60–80% of the epithelium throughout the tubule [6,12], while basal cells make up 6–30% [13,14]. Narrow and apical cells are predominantly found in the initial segment of the epididymis, whereas other cell types are distributed throughout the entire epididymal tissue [8,11,15,16,17]. The detailed functions and protein markers of the epithelial cells are listed in Table 1. In addition, a small number of non-epithelial cells, such as macrophages/monocytes, mononuclear phagocytes, and T lymphocytes, are shown [18,19,20,21,22,23].
The in vitro culture of the epididymal epithelium is frequently employed to study the role of the epididymis in sperm maturation and the associated molecular mechanisms. In 1986, studies reported that epididymal epithelial cells were successfully isolated from adult rats and exhibited polarized characteristics only when plated at high densities (>1 × 106 cell/cm2) [31]. By 1990, a methodology was introduced that allowed for the sustained culture of human epididymal cells over a period of 42 days, marking a significant advancement [32]. Epididymal epithelial cells from various species can be isolated and cultivated successfully in vitro, with some demonstrating functional capacities, particularly in promoting sperm maturation and enhancing sperm motility [33,34,35,36,37,38,39]. Despite extensive research into the relationship between epididymal epithelial cells and sperm maturation, the underlying molecular mechanisms remain poorly understood. This gap may be attributed to the inability of monolayer cultures or in vitro-passaged cells to faithfully replicate the complex biology of epididymal epithelial cells acquired in vivo, thus limiting our understanding of these intricate biological processes. Epithelial principal cell lines derived from humans [40,41], rats [42], and mice [43,44] have been employed to investigate the role of cellular communication in the epididymis [45,46,47], and to assess reproductive toxicity [47]. However, these cell lines originate from a singular source and do not fully represent the comprehensive biological functions of epididymal tissue.
Tissue culture models have successfully elucidated aspects of epididymal biology and supported drug toxicity testing [48,49,50,51,52]. Although the in vitro culture model of epididymal tissue provides a powerful platform for studying the function of the epididymis in various species [48,53], it remains operationally challenging. Previous research has demonstrated that the isolation and cultivation of primary human epididymal epithelial cells are relatively well established [54,55]. However, the in vitro culture of rodent epididymal monolayer epithelial cells has rarely been reported. Although there are a few reports, normal morphology in the culture conditions has proved difficult to maintain [56,57]. Given the diverse cellular composition of the epididymal epithelium, a 3D in vitro model provides a more comprehensive and realistic approach to investigating and understanding the intricate facets of epididymal function.
Organoids are three-dimensional structures composed of multiple cells that closely mimic the cellular structure and function of organs [58]. This similarity in structure and function facilitates the investigation of complex cell interactions and tissue development processes [59]. The emergence of organoid research has opened new avenues for both fundamental and translational research over the past decade [60]. Organoids, such as testis organoids, hold significant promise in reproductive biology and toxicology, whether in animal or human models [61,62,63]. Similarly, epididymal organoids also exhibit considerable potential. Recent in vitro studies have successfully demonstrated the formation of epididymal organoids from single-cell suspensions in different species. This review highlights recent advances in the generation of epididymal organoids and their potential applications.

2. The Main Function of the Epididymal Epithelium

The distribution and characteristics of the epididymal epithelium are crucial for sperm maturation, with epithelial cells exhibiting varied morphologies and functions across different regions. Sperm are generated in the seminiferous tubules of the testis. Initially devoid of motility and fertilization capabilities, spermatozoa acquire these functions during transit in the epididymal lumen—a process known as sperm maturation [9,64,65]. The components of the epididymal luminal fluid are mainly synthesized and secreted by various types of epithelial cells lining the duct [66]. The luminal fluid in the caput and corpus of the epididymis can assist sperm in acquiring motility and fertilization ability, while the luminal fluid in the cauda is beneficial for sperm storage [67]. Epididymal epithelial cells play a crucial role in establishing a highly specialized luminal microenvironment. This microenvironment is specifically tailored to promote a gradient that enhances the fertility of the sperm population contained within [68]. Despite the fact that the complexity of this process presents challenges and it lacks full comprehension, specific facets of sperm maturation have been firmly established [6,69,70].
Multiple factors are responsible for sperm maturation in the epididymal lumen. These include proteins secreted by the principal cells in the epididymis that bind to maturing spermatozoa; exosomes released by the apical plasma, called epididymal exosomes, which transport cargo to the sperm; and pH fluctuations throughout the epididymis [67,71,72,73]. The blood–epididymal barrier (BEB) is formed by apical tight junctions between the principal cells, enabling the selective transportation of molecules through the epithelium. These tight junctions consist of integral proteins that play a central role in determining the barrier’s selective permeability, thereby creating the luminal environment conducive to facilitating sperm maturation [6,74]. The maintenance of a normal epididymal epithelium is indispensable for proper sperm maturation, and epididymal dysfunction is intricately linked to infertility [40,41,75,76].
The formation of organoids in the epididymis holds great promise for revealing the underlying molecular mechanisms that regulate epididymal function. However, a crucial prerequisite for the development of epididymal organoids is the presence of stem cells or progenitor cells within the epididymis [77,78]. Are there stem cells or progenitor cells in the epididymis that can give rise to these organoids?

3. Basal Cell—The Prerequisite for Organoid Formation?

As early as 1925, researchers using a rat model first suggested the existence of stem cells within the epididymal epithelium [79]. Subsequent hypotheses proposed basal cells as potential stem cells in the epididymis [80,81]. Observations in unilaterally orchiectomized adult male rats revealed that basal cells exhibited a transition from an oval to a triangular and elongated shape, evolving into expanded columnar cells [79]. In vitro studies demonstrated that basal cells, identified by keratin 5 (KRT5) positivity, could differentiate into cells expressing KRT8 and connexin 26, markers typical of columnar cells [13]. These basal cells showed self-renewal and differentiation capabilities, forming organoids capable of expressing aquaporin 9 and CFTR, indicative of principal cell markers [82,83]. Furthermore, these cells secreted clusterin, a protein crucial for spermatozoa maturation [84]. Basal-cell-derived organoids exhibited self-renewal potential, maintaining newly formed organoids for at least 13 passages [84]. This evidence strongly supports the characterization of basal cells as possessing stem cell-like properties with significant self-renewal capacity [85]. Previous research has documented segment-specific gene expression and regulation within the epididymis [86,87]. Moreover, gene expression profiles of the principal cells varied between the proximal and distal segments. Interestingly, no significant differences were observed in the organoids derived from basal cells isolated from either proximal or distal epididymal regions [84]. This suggests that regional differences in gene expression may not originate solely from the specific segmental origin of basal cells.
In the epididymis, GJB2 serves as a marker for columnar cells, with its expression levels decreasing significantly as these cells differentiate into principal and other cell types [88]. GJB2 was not detected in basal cells cultured in vitro for 3 days; however, its expression became evident in cells within the acini after 7, 10, and 14 days of culture [13]. This suggests that basal cells possess the capacity to differentiate into cells resembling columnar cells. A similar mechanism has been observed in the trachea, where exposure to SO2 depletes ciliated cells, prompting basal cells to differentiate initially into undifferentiated progenitors. These progenitors then progress through differentiation stages to become ciliated and secretory cells, indicating a sequential two-step differentiation process [89]. Therefore, this implies the feasibility of regenerating the epididymal epithelium, potentially shedding light on the adaptability of this crucial organ in male fertility. Upon single-cell analysis, three distinct clusters of basal cells were identified, demonstrating the common expression of marker genes such as Itga6 and Krt14. These clusters exhibited an enrichment of genes mainly involved in cell adhesion, membrane transport, and lipid metabolism [86]. The precise nature of these basal cell clusters—whether they represent distinct cell types, different stages of differentiation, or separate adult stem cell populations—remains ambiguous. Accordingly, the selection of KRT5-positive or ITGA6-positive cells may have biased the enrichment towards specific basal cell subpopulations [90].
On the contrary, other studies have indicated age-related characteristics in basal cells, which challenge their classification as stem cells [91]. In contrast, organs like the liver and amniotic membrane have exhibited epithelia containing expanding stem cells [92,93]. Additionally, quiescent adult stem cells with active regenerative properties have been identified in many tissues [94], such as the salivary gland [95], liver [96], intestine [97], and pituitary [98]. Therefore, although the low proliferation or expansion index of epididymal epithelial cells does not conclusively prove the existence of stem cells in the epididymis [79], ongoing debate persists regarding whether basal cells in the epididymis fulfill the criteria of adult stem cells [99]. Even without confirmation as true stem cells, basal cells likely retain differentiation potential and contribute to organoid formation [84]. Thus, despite ongoing controversy, researchers can continue to employ basal cells from the epididymis for in vitro culture and organoid studies.

4. Development History of Epididymal Organoids

A well-designed microenvironment in tissue and cell engineering can promote proliferation, migration, matrix production, and stem cell differentiation. Significant differences exist regarding cell–cell interactions, cellular mechanics, and nutrient access between 3D and standard 2D cell cultures, as noted by reference [100]. Nevertheless, 2D monolayer cell culture systems may not accurately simulate the observed cell development process in the in vivo physiological environment due to their inherent simplicity. This discrepancy stems from the lack of a complex, biologically rich environment. The advent of 3D cell culture approaches, which model in vivo tissue and organ interactions, has opened new avenues for studying underlying biochemical and biomechanical signals [101,102]. Given their ability to more closely mimic the in vivo environment, 3D culture systems are gaining popularity. The term “organoids” was coined in 1947 within the field of oncology [103]. With advancements in stem cell biotechnology, particularly the refinement of three-dimensional (3D) cell culture techniques, the definition of organoids has evolved to encompass 3D in vitro structures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs), exhibiting near-native microanatomy [78,104].
The first reported case of organoids used intestinal cells, which was published in Nature in 2007, and marked a significant breakthrough in biological research. These studies identified leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) as a specific marker gene for intestinal stem cells, enabling their characterization and purification [105]. Subsequent research revealed the capacity of adult intestinal stem cells to proliferate and differentiate both in vivo and in vitro [106,107]. These findings underscored the potential of 3D culture techniques to support ASC self-renewal and the formation of organ-like structures, thus offering promising avenues for tissue regeneration research. Since then, organoids have been successfully developed from various tissues including the stomach [108], liver [109,110], brain [111], prostate [112], mammary gland [113], testis [114,115,116], endometrium [117], fallopian tube [118,119], ovary [120], and epididymis [84,121,122,123], among others [60,124,125].
The development of 3D culture technologies has enabled the use of in vitro models to study epididymal function mechanisms [39,99,126]. Early epididymal structures have been observed to form epididymal spheroids under both 2D and 3D conditions [13,34,127,128]. This review provides an overview of the developmental history of epididymal spheroids or organoids cultivated in various species (see Figure 2). The formation process of epididymal organoids involves digesting epididymal tissue into single cells, after which the epithelial cells or basal cells within the epididymal tissue can spontaneously re-aggregate to form spheres and organoid structures, resembling the arrangement of epithelial cells in vitro [8,121]. More detailed information is summarized in Table 2.
Mou et al. first reported epididymal organoids in mice in 2016, isolating KRT5-positive basal cells to construct organoids consisting of basal and clear cells in vitro. When these basal cells were subcutaneously injected into nude mice, they differentiated and formed spherical structures comprising basal and principal cells [99]. The matrix is typically composed of the ECM (extracellular matrix), and the cell density can be set at 2 × 104 cells. The basic culture medium utilizes DMEM/F12 supplemented with 25 ng/mL of EGF, and the culture cycle can extend up to 10 days. Pinel and Cyr isolated basal cells from rats and cultured epididymis-like organoids in vitro, highlighting the stem cell characteristics of basal cells. The organoids were cultured by depositing homogeneous cell suspensions as 50 μL drops onto Matrigel-coated, 24-well plates and incubating them upside down at 37 °C for 30 min to solidify the Matrigel [84]. Dufresne et al., from the same research group, utilized rat epididymis organoids to simulate epididymal development and analyze gene expression profiles through transcriptomic analysis across different stages of organoid growth [123]. These studies predominantly focused on epididymal organoids derived from caput, proximal, or distal epididymal regions, demonstrating their self-assembly and differentiation capabilities.
Based on the aforementioned studies, we extended our research to construct epididymal organoids from different regions, specifically the caput, corpus, and cauda, and analyzed their respective gene expression profiles. In our laboratory, and based on the methods used for generating human [121] and rat [84] organoids, we successfully optimized a protocol for the formation of mouse epididymis organoids, as illustrated in Figure 3. We obtained epithelial cells derived from different regions of the mouse epididymis and successfully generated organoids resembling the caput, corpus, and cauda of the epididymis in vitro. We mixed the appropriate concentration of basement membrane extract (BME) with a sufficient quantity of epididymal epithelial cells and supplemented this with EGF, testosterone, dihydrotestosterone, retinoic acid, and other additive factors. We took 10 μL of the mixed cell suspension at a low temperature and created a small droplet in a 96-well cell culture plate. Once the droplet solidified, and with the plate the right way up, we added an appropriate volume of organoid culture medium until the organoid formed. This method is much simpler than those previously reported [84].

5. Potential Application of Epididymal Organoids

Organoid models derived from the cells of mouse, rat, and human epididymis have been established [121,122,128]. These novel in vitro cell culture systems represent a significant advancement complementing existing epididymal cell lines and animal models. Organoids are pivotal for advancing the precision of male infertility treatment [127,129]. Similar to testicular organoids in male reproduction, epididymal organoids offer a valuable platform for the high-throughput screening of drugs and toxicity [130], although research in this area is still in its developmental stage. Limited knowledge suggests a paucity of information regarding the drug screening and clinical applications of epididymal organoids. The majority of research efforts have been directed towards establishing culture systems and studying the simulation of epididymal development. Drawing from research on organoids derived from other tissues [59,131,132,133], potential applications of epididymal organoids could include the following:
(1) Disease modeling: Epididymal organoids offer a promising avenue for disease modeling and studying various epididymal conditions such as infertility and congenital abnormalities. In males with cystic fibrosis, defects in the epididymis or vas deferens often lead to obstructive azoospermia [134]. Leir et al. utilized a human epididymal epithelial cell organoid model to elucidate the molecular mechanisms underlying male infertility mediated by CFTR in cystic fibrosis patients [121]. Thus, epididymal organoids demonstrate significant potential for studying male infertility and for screening therapeutic drugs. (2) Drug testing: Organoids cultured from prostate cancer patients have demonstrated resistance to cell growth arrest and apoptosis induced by BET inhibitors, revealing a new molecular mechanism for BET inhibitor resistance in these patients [135,136]. Recent epidemics, such as the COVID-19 pandemic, have been associated with impaired sperm in males with moderate SARS-CoV-2 infection [137,138]. The development of testis and epididymis-like organs can offer a more suitable ex vivo model for studying such acute events. Testis organoids have emerged as effective tools for organ-level reproductive toxicity screening [63,114,139], yet the role of epididymal organoids in drug screening and virus infection treatment remains unexplored. Thus, establishing epididymal models for drug screening is crucial to advance research on viral resistance in reproductive organs. (3) Reproductive biology research: Organoids can partly replicate the structural and functional characteristics of in vivo organs, facilitating the study of cell interactions and signaling pathways through organoid cultures [8,58]. Early studies indicated that epididymal epithelial cells play a role in promoting sperm maturation, offering a platform for investigating this process in vitro [34,35,37]. However, the limitations of 2D culture conditions hinder a comprehensive exploration of how the epididymal epithelium impacts sperm maturation and its underlying mechanisms [36,140,141]. Organoid technology allows researchers to manipulate cell types and observe their re-aggregation dynamics, a capability not feasible in traditional organotypic cultures [142]. At present, there is no report on whether epididymal organoids promote sperm maturation, which could be one of the directions of future research.

6. Conclusions

Until now, only a limited number of 3D epididymal organoids have been developed [84]. Several challenges are associated with creating 3D organoids of the epididymis. An accurate simulation of the diverse segments of the epididymis would be advantageous for studying sperm transport, maturation, acquisition of motility, and their underlying molecular mechanisms. Researchers have encountered difficulties due to the scarcity of epididymal tissue samples and the challenges involved in obtaining tissue from younger patients who do not have epididymal cancer. The advent of the male reproductive system organoids, encompassing prostatic, testicular, epididymal, and potentially seminal vesicle organoids, suggests a promising trajectory towards their integration into a cohesive, multi-organ-on-a-chip platform. Moreover, epididymal organoids serve as a valuable tool for elucidating infertility mechanisms, studying treatment efficacy, and evaluating drug toxicity. Future investigations should focus on refining them, with a critical need to elucidate the physiological and pathological contexts related to these changes. This understanding is pivotal for elucidating their implications in both clinical and physiological studies.

Author Contributions

J.N. and H.C. conceived the study. X.Z. and J.N. wrote and polished the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by Startup R&D funding from Nantong University (135420631138 to Junyu Nie), the Natural Science Program of Nantong University School of Medicine (TDYX2021015 to Junyu Nie), and the Natural Science Foundation of Jiangsu Province (BK20210840 to Xiuling Zhao).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Jelinsky, S.A.; Turner, T.T.; Bang, H.J.; Finger, J.N.; Solarz, M.K.; Wilson, E.; Brown, E.L.; Kopf, G.S.; Johnston, D.S. The rat epididymal transcriptome: Comparison of segmental gene expression in the rat and mouse epididymides. Biol. Reprod. 2007, 76, 561–570. [Google Scholar] [CrossRef]
  2. Johnston, D.S.; Turner, T.T.; Finger, J.N.; Owtscharuk, T.L.; Kopf, G.S.; Jelinsky, S.A. Identification of epididymis-specific transcripts in the mouse and rat by transcriptional profiling. Asian J. Androl. 2007, 9, 522–527. [Google Scholar] [CrossRef]
  3. Legare, C.; Akintayo, A.; Blondin, P.; Calvo, E.; Sullivan, R. Impact of male fertility status on the transcriptome of the bovine epididymis. Mol. Hum. Reprod. 2017, 23, 355–369. [Google Scholar] [CrossRef] [PubMed]
  4. Turner, T.T.; Bomgardner, D.; Jacobs, J.P.; Nguyen, Q.A. Association of segmentation of the epididymal interstitium with segmented tubule function in rats and mice. Reproduction 2003, 125, 871–878. [Google Scholar] [CrossRef] [PubMed]
  5. Gong, Q.Q.; Dou, Z.L.; Wang, X.; Zhang, K.Y.; Chen, H.; Gao, J.G.; Sun, X.Y. Epididymal initial segment-specific Cre recombinase activity in Lcn8-Cre knock-in mice. Mol. Biol. Rep. 2021, 48, 6015–6023. [Google Scholar] [CrossRef] [PubMed]
  6. Cornwall, G.A. New insights into epididymal biology and function. Hum. Reprod. Update 2009, 15, 213–227. [Google Scholar] [CrossRef] [PubMed]
  7. Elbashir, S.; Magdi, Y.; Rashed, A.; Henkel, R.; Agarwal, A. Epididymal contribution to male infertility: An overlooked problem. Andrologia 2021, 53, e13721. [Google Scholar] [CrossRef]
  8. Dufresne, J.; Gregory, M.; Pinel, L.; Cyr, D.G. Three-Dimensional Cell Culture of Epididymal Basal Cells and Organoids: A Novel Tool for Toxicology. Curr. Protoc. 2024, 4, e975. [Google Scholar] [CrossRef]
  9. Sullivan, R.; Legare, C.; Lamontagne-Proulx, J.; Breton, S.; Soulet, D. Revisiting structure/functions of the human epididymis. Andrology 2019, 7, 748–757. [Google Scholar] [CrossRef]
  10. Breton, S.; Ruan, Y.C.; Park, Y.J.; Kim, B. Regulation of epithelial function, differentiation, and remodeling in the epididymis. Asian J. Androl. 2016, 18, 3–9. [Google Scholar] [CrossRef]
  11. Shum, W.W.; Ruan, Y.C.; Da Silva, N.; Breton, S. Establishment of cell-cell cross talk in the epididymis: Control of luminal acidification. J. Androl. 2011, 32, 576–586. [Google Scholar] [CrossRef] [PubMed]
  12. Ma, W.; Li, S.; Ma, S.; Jia, L.; Zhang, F.; Zhang, Y.; Zhang, J.; Wong, G.; Zhang, S.; Lu, X.; et al. Zika Virus Causes Testis Damage and Leads to Male Infertility in Mice. Cell 2016, 167, 1511–1524.e10. [Google Scholar] [CrossRef] [PubMed]
  13. Mandon, M.; Hermo, L.; Cyr, D.G. Isolated Rat Epididymal Basal Cells Share Common Properties with Adult Stem Cells. Biol. Reprod. 2015, 93, 115. [Google Scholar] [CrossRef] [PubMed]
  14. Seiler, P.; Wenzel, I.; Wagenfeld, A.; Yeung, C.H.; Nieschlag, E.; Cooper, T.G. The appearance of basal cells in the developing murine epididymis and their temporal expression of macrophage antigens. Int. J. Androl. 1998, 21, 217–226. [Google Scholar] [CrossRef] [PubMed]
  15. Jégou, B.; Skinner, M.K. Content and Volume Overview; Academic Press: New York, NY, USA, 2018; pp. 1–2. [Google Scholar]
  16. Adamali, H.I.; Hermo, L. Apical and narrow cells are distinct cell types differing in their structure, distribution, and functions in the adult rat epididymis. J. Androl. 1996, 17, 208–222. [Google Scholar] [CrossRef] [PubMed]
  17. Belleannee, C.; Thimon, V.; Sullivan, R. Region-specific gene expression in the epididymis. Cell Tissue Res. 2012, 349, 717–731. [Google Scholar] [CrossRef] [PubMed]
  18. Battistone, M.A.; Elizagaray, M.L.; Barrachina, F.; Ottino, K.; Mendelsohn, A.C.; Breton, S. Immunoregulatory mechanisms between epithelial clear cells and mononuclear phagocytes in the epididymis. Andrology 2023, 12, 949–963. [Google Scholar] [CrossRef] [PubMed]
  19. Battistone, M.A.; Mendelsohn, A.C.; Spallanzani, R.G.; Brown, D.; Nair, A.V.; Breton, S. Region-specific transcriptomic and functional signatures of mononuclear phagocytes in the epididymis. Mol. Hum. Reprod. 2020, 26, 14–29. [Google Scholar] [CrossRef]
  20. Breton, S.; Nair, A.V.; Battistone, M.A. Epithelial dynamics in the epididymis: Role in the maturation, protection, and storage of spermatozoa. Andrology 2019, 7, 631–643. [Google Scholar] [CrossRef]
  21. Shum, W.W.; Hill, E.; Brown, D.; Breton, S. Plasticity of basal cells during postnatal development in the rat epididymis. Reproduction 2013, 146, 455–469. [Google Scholar] [CrossRef]
  22. Voisin, A.; Damon-Soubeyrand, C.; Bravard, S.; Saez, F.; Drevet, J.R.; Guiton, R. Differential expression and localisation of TGF-beta isoforms and receptors in the murine epididymis. Sci. Rep. 2020, 10, 995. [Google Scholar] [CrossRef]
  23. Shum, W.W.; Smith, T.B.; Cortez-Retamozo, V.; Grigoryeva, L.S.; Roy, J.W.; Hill, E.; Pittet, M.J.; Breton, S.; Da Silva, N. Epithelial basal cells are distinct from dendritic cells and macrophages in the mouse epididymis. Biol. Reprod. 2014, 90, 90. [Google Scholar] [CrossRef] [PubMed]
  24. Chen, H.; Alves, M.B.R.; Belleannee, C. Contribution of epididymal epithelial cell functions to sperm epigenetic changes and the health of progeny. Hum. Reprod. Update 2022, 28, 51–66. [Google Scholar] [CrossRef] [PubMed]
  25. Belleannee, C.; Da Silva, N.; Shum, W.W.; Marsolais, M.; Laprade, R.; Brown, D.; Breton, S. Segmental expression of the bradykinin type 2 receptor in rat efferent ducts and epididymis and its role in the regulation of aquaporin 9. Biol. Reprod. 2009, 80, 134–143. [Google Scholar] [CrossRef] [PubMed]
  26. Sharma, S.; Kumaran, G.K.; Hanukoglu, I. High-resolution imaging of the actin cytoskeleton and epithelial sodium channel, CFTR, and aquaporin-9 localization in the vas deferens. Mol. Reprod. Dev. 2020, 87, 305–319. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, M.M.; Feng, X.L.; Qi, C.; Zhang, S.E.; Zhang, G.L. The significance of single-cell transcriptome analysis in epididymis research. Front. Cell Dev. Biol. 2024, 12, 1357370. [Google Scholar] [CrossRef]
  28. Isnard-Bagnis, C.; Da Silva, N.; Beaulieu, V.; Yu, A.S.; Brown, D.; Breton, S. Detection of ClC-3 and ClC-5 in epididymal epithelium: Immunofluorescence and RT-PCR after LCM. Am. J. Physiol. Cell Physiol. 2003, 284, C220–C232. [Google Scholar] [CrossRef] [PubMed]
  29. Martinez-Garcia, F.; Regadera, J.; Cobo, P.; Palacios, J.; Paniagua, R.; Nistal, M. The apical mitochondria-rich cells of the mammalian epididymis. Andrologia 1995, 27, 195–206. [Google Scholar] [CrossRef]
  30. Leir, S.H.; Yin, S.; Kerschner, J.L.; Cosme, W.; Harris, A. An atlas of human proximal epididymis reveals cell-specific functions and distinct roles for CFTR. Life Sci. Alliance 2020, 3, 11. [Google Scholar] [CrossRef]
  31. Byers, S.W.; Hadley, M.A.; Djakiew, D.; Dym, M. Growth and characterization of polarized monolayers of epididymal epithelial cells and Sertoli cells in dual environment culture chambers. J. Androl. 1986, 7, 59–68. [Google Scholar] [CrossRef]
  32. Cooper, T.G.; Yeung, C.H.; Meyer, R.; Schulze, H. Maintenance of human epididymal epithelial cell function in monolayer culture. J. Reprod. Fertil. 1990, 90, 81–91. [Google Scholar] [CrossRef] [PubMed]
  33. Moore, H.D.; Hartman, T.D.; Smith, C.A. In-vitro culture of hamster epididymal epithelium and induction of sperm motility. J. Reprod. Fertil. 1986, 78, 327–336. [Google Scholar] [CrossRef] [PubMed]
  34. Moore, H.D.; Curry, M.R.; Penfold, L.M.; Pryor, J.P. The culture of human epididymal epithelium and in vitro maturation of epididymal spermatozoa. Fertil. Steril. 1992, 58, 776–783. [Google Scholar] [CrossRef] [PubMed]
  35. Kervancioglu, M.E.; Djahanbakhch, O.; Aitken, R.J. Epithelial cell coculture and the induction of sperm capacitation. Fertil. Steril. 1994, 61, 1103–1108. [Google Scholar] [CrossRef] [PubMed]
  36. Bongso, A.; Trounson, A. Evaluation of motility, freezing ability and embryonic development of murine epididymal sperm after coculture with epididymal epithelium. Hum. Reprod. 1996, 11, 1451–1456. [Google Scholar] [CrossRef] [PubMed]
  37. Akhondi, M.A.; Chapple, C.; Moore, H.D. Prolonged survival of human spermatozoa when co-incubated with epididymal cell cultures. Hum. Reprod. 1997, 12, 514–522. [Google Scholar] [CrossRef] [PubMed]
  38. Lin, M.; Zhang, X.; Murdoch, R.; Aitken, R.J. In vitro culture of brushtail possum (Trichosurus vulpecula) epididymal epithelium and induction of epididymal sperm maturation in co-culture. J. Reprod. Fertil. 2000, 119, 1–14. [Google Scholar] [CrossRef] [PubMed]
  39. Wei, Y.S.; Lin, W.Z.; Wang, T.E.; Lee, W.Y.; Li, S.H.; Lin, F.J.; Nixon, B.; Sipila, P.; Tsai, P.S. Polarized epithelium-sperm co-culture system reveals stimulatory factors for the secretion of mouse epididymal quiescin sulfhydryl oxidase 1. J. Reprod. Dev. 2022, 68, 198–208. [Google Scholar] [CrossRef] [PubMed]
  40. Dube, E.; Dufresne, J.; Chan, P.T.; Hermo, L.; Cyr, D.G. Assessing the role of claudins in maintaining the integrity of epididymal tight junctions using novel human epididymal cell lines. Biol. Reprod. 2010, 82, 1119–1128. [Google Scholar] [CrossRef]
  41. Dube, E.; Hermo, L.; Chan, P.T.; Cyr, D.G. Alterations in the human blood-epididymis barrier in obstructive azoospermia and the development of novel epididymal cell lines from infertile men. Biol. Reprod. 2010, 83, 584–596. [Google Scholar] [CrossRef]
  42. Dufresne, J.; St-Pierre, N.; Viger, R.S.; Hermo, L.; Cyr, D.G. Characterization of a novel rat epididymal cell line to study epididymal function. Endocrinology 2005, 146, 4710–4720. [Google Scholar] [CrossRef] [PubMed]
  43. Araki, Y.; Suzuki, K.; Matusik, R.J.; Obinata, M.; Orgebin-Crist, M.C. Immortalized epididymal cell lines from transgenic mice overexpressing temperature-sensitive simian virus 40 large T-antigen gene. J. Androl. 2002, 23, 854–869. [Google Scholar] [CrossRef] [PubMed]
  44. Sipila, P.; Shariatmadari, R.; Huhtaniemi, I.T.; Poutanen, M. Immortalization of epididymal epithelium in transgenic mice expressing simian virus 40 T antigen: Characterization of cell lines and regulation of the polyoma enhancer activator 3. Endocrinology 2004, 145, 437–446. [Google Scholar] [CrossRef] [PubMed]
  45. Adam, C.; Cyr, D.G. Role of Specificity Protein-1 and Activating Protein-2 Transcription Factors in the Regulation of the Gap Junction Protein Beta-2 Gene in the Epididymis of the Rat. Biol. Reprod. 2016, 94, 120. [Google Scholar] [CrossRef] [PubMed]
  46. Dufresne, J.; Cyr, D.G. Activation of an SP binding site is crucial for the expression of claudin 1 in rat epididymal principal cells. Biol. Reprod. 2007, 76, 825–832. [Google Scholar] [CrossRef] [PubMed]
  47. Jones, S.R.; Cyr, D.G. Regulation and characterization of the ATP-binding cassette transporter-B1 in the epididymis and epididymal spermatozoa of the rat. Toxicol. Sci. 2011, 119, 369–379. [Google Scholar] [CrossRef] [PubMed]
  48. Ellerbrock, K.; Pera, I.; Hartung, S.; Ivell, R. Gene expression in the dog epididymis: A model for human epididymal function. Int. J. Androl. 1994, 17, 314–323. [Google Scholar] [CrossRef] [PubMed]
  49. Pearl, C.A.; Roser, J.F. Lactoferrin expression and secretion in the stallion epididymis. Reprod. Biol. 2014, 14, 148–154. [Google Scholar] [CrossRef] [PubMed]
  50. Kumar, M.; Tanwar, P. Organ Culture and Whole Mount Immunofluorescence Staining of Mouse Wolffian Ducts. J. Vis. Exp. 2017, 119, e55134. [Google Scholar]
  51. Klinefelter, G.R.; Hamilton, D.W. Organ culture of rat caput epididymal tubules in a perifusion chamber. J. Androl. 1984, 5, 243–258. [Google Scholar] [CrossRef]
  52. Battaglia, G. Preliminary observations on movements of the epididymis of the rat in rotating organ type culture. Boll. Soc. Ital. Biol. Sper. 1956, 32, 265–267. [Google Scholar] [PubMed]
  53. Kaur, J.; Ramakrishnan, P.R.; Rajalakshmi, M. In vitro organ culture of rhesus monkey epididymal tubules. Contraception 1991, 43, 295–303. [Google Scholar] [CrossRef] [PubMed]
  54. Leir, S.H.; Browne, J.A.; Eggener, S.E.; Harris, A. Characterization of primary cultures of adult human epididymis epithelial cells. Fertil. Steril. 2015, 103, 647–654.e1. [Google Scholar] [CrossRef] [PubMed]
  55. Coatti, G.C.; Paranjapye, A.; Harris, A. Dual SMAD inhibition enhances the longevity of human epididymis epithelial cells. Cell Tissue Res. 2023, 391, 409–417. [Google Scholar] [CrossRef] [PubMed]
  56. Buff, S.; Lambert, V.; Marchal, T.; Guerin, P. Isolation, culture and characteristics of epididymal epithelial cells from adult cats. Theriogenology 2005, 64, 1603–1618. [Google Scholar] [CrossRef] [PubMed]
  57. Qu, B.; Gu, Y.; Shen, J.; Qin, J.; Bao, J.; Hu, Y.; Zeng, W.; Dong, W. Trehalose maintains vitality of mouse epididymal epithelial cells and mediates gene transfer. PLoS ONE 2014, 9, e92483. [Google Scholar] [CrossRef]
  58. Jiang, Y.; Zhao, H.; Kong, S.; Zhou, D.; Dong, J.; Cheng, Y.; Zhang, S.; Wang, F.; Kalra, A.; Yang, N.; et al. Establishing mouse and human oral esophageal organoids to investigate the tumor immune response. Dis. Model. Mech. 2024, 17, dmm050319. [Google Scholar] [CrossRef] [PubMed]
  59. Smirnov, A.; Melino, G.; Candi, E. Gene expression in organoids: An expanding horizon. Biol. Direct 2023, 18, 11. [Google Scholar] [CrossRef]
  60. Corro, C.; Novellasdemunt, L.; Li, V.S.W. A brief history of organoids. Am. J. Physiol. Cell Physiol. 2020, 319, C151–C165. [Google Scholar] [CrossRef] [PubMed]
  61. Tang, S.; Jones, C.; Mecca, R.; Davies, J.; Lane, S.; Coward, K. Anin vitrothree-dimensional (3D) testicular organoid culture system for efficient gonocyte maintenance and propagation using frozen/thawed neonatal bovine testicular tissues. Biomed. Mater. 2024, 19, 025040. [Google Scholar] [CrossRef]
  62. Richer, G.; Vanhaecke, T.; Rogiers, V.; Goossens, E.; Baert, Y. Mouse In Vitro Spermatogenesis on 3D Bioprinted Scaffolds. Methods Mol. Biol. 2024, 2770, 135–149. [Google Scholar] [PubMed]
  63. Skardal, A.; Aleman, J.; Forsythe, S.; Rajan, S.; Murphy, S.; Devarasetty, M.; Pourhabibi Zarandi, N.; Nzou, G.; Wicks, R.; Sadri-Ardekani, H.; et al. Drug compound screening in single and integrated multi-organoid body-on-a-chip systems. Biofabrication 2020, 12, 025017. [Google Scholar] [CrossRef] [PubMed]
  64. Bedford, J.M.; Calvin, H.; Cooper, G.W. The maturation of spermatozoa in the human epididymis. J. Reprod. Fertil. Suppl. 1973, 18, 199–213. [Google Scholar] [PubMed]
  65. Bedford, J.M. The status and the state of the human epididymis. Hum. Reprod. 1994, 9, 2187–2199. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, X.; Qiu, F.; Yu, J.; Zhou, M.; Zuo, A.; Xu, X.; Sun, X.Y.; Wang, Z. Transcriptome profiling of the initial segment and proximal caput of mouse epididymis. Front. Endocrinol. 2023, 14, 1190890. [Google Scholar] [CrossRef] [PubMed]
  67. Barrachina, F.; Battistone, M.A.; Castillo, J.; Mallofre, C.; Jodar, M.; Breton, S.; Oliva, R. Sperm acquire epididymis-derived proteins through epididymosomes. Hum. Reprod. 2022, 37, 651–668. [Google Scholar] [CrossRef] [PubMed]
  68. Turner, T.T. Resorption versus secretion in the rat epididymis. J. Reprod. Fertil. 1984, 72, 509–514. [Google Scholar] [CrossRef] [PubMed]
  69. Turner, T.T. On the epididymis and its role in the development of the fertile ejaculate. J. Androl. 1995, 16, 292–298. [Google Scholar] [CrossRef] [PubMed]
  70. Sullivan, R.; Mieusset, R. The human epididymis: Its function in sperm maturation. Hum. Reprod. Update 2016, 22, 574–587. [Google Scholar] [CrossRef] [PubMed]
  71. Breton, S.; Brown, D. Regulation of luminal acidification by the V-ATPase. Physiology 2013, 28, 318–329. [Google Scholar] [CrossRef]
  72. Sharma, U.; Conine, C.C.; Shea, J.M.; Boskovic, A.; Derr, A.G.; Bing, X.Y.; Belleannee, C.; Kucukural, A.; Serra, R.W.; Sun, F.; et al. Biogenesis and function of tRNA fragments during sperm maturation and fertilization in mammals. Science 2016, 351, 391–396. [Google Scholar] [CrossRef] [PubMed]
  73. Sullivan, R. Epididymosomes: A heterogeneous population of microvesicles with multiple functions in sperm maturation and storage. Asian J. Androl. 2015, 17, 726–729. [Google Scholar] [CrossRef] [PubMed]
  74. Dube, E.; Cyr, D.G. The blood-epididymis barrier and human male fertility. Adv. Exp. Med. Biol. 2012, 763, 218–236. [Google Scholar] [PubMed]
  75. Mital, P.; Hinton, B.T.; Dufour, J.M. The blood-testis and blood-epididymis barriers are more than just their tight junctions. Biol. Reprod. 2011, 84, 851–858. [Google Scholar] [CrossRef] [PubMed]
  76. Liu, W.H.; Wang, F.; Yu, X.Q.; Wu, H.; Gong, M.L.; Chen, R.; Zhang, W.J.; Han, R.Q.; Liu, A.J.; Chen, Y.M.; et al. Damaged male germ cells induce epididymitis in mice. Asian J. Androl. 2020, 22, 472–480. [Google Scholar] [PubMed]
  77. Rossi, G.; Manfrin, A.; Lutolf, M.P. Progress and potential in organoid research. Nat. Rev. Genet. 2018, 19, 671–687. [Google Scholar] [CrossRef] [PubMed]
  78. Kretzschmar, K.; Clevers, H. Organoids: Modeling Development and the Stem Cell Niche in a Dish. Dev. Cell 2016, 38, 590–600. [Google Scholar] [CrossRef] [PubMed]
  79. Pinel, L.; Mandon, M.; Cyr, D.G. Tissue regeneration and the epididymal stem cell. Andrology 2019, 7, 618–630. [Google Scholar] [CrossRef] [PubMed]
  80. Hamilton, D.W. Structure and function of the epithelium lining the ductuli efferentes, ductus epididymidis and ductus deferens in the rat. In Handbook of Physiology Endocrinology; American Physiological Society: Washington, DC, USA, 1975. [Google Scholar]
  81. Ramos, A.S., Jr.; Dym, M. Fine structure of the monkey epididymis. Am. J. Anat. 1977, 149, 501–531. [Google Scholar] [CrossRef]
  82. Ruan, Y.C.; Shum, W.W.; Belleannee, C.; Da Silva, N.; Breton, S. ATP secretion in the male reproductive tract: Essential role of CFTR. J. Physiol. 2012, 590, 4209–4222. [Google Scholar] [CrossRef]
  83. Pastor-Soler, N.; Bagnis, C.; Sabolic, I.; Tyszkowski, R.; McKee, M.; Van Hoek, A.; Breton, S.; Brown, D. Aquaporin 9 expression along the male reproductive tract. Biol. Reprod. 2001, 65, 384–393. [Google Scholar] [CrossRef] [PubMed]
  84. Pinel, L.; Cyr, D.G. Self-renewal and differentiation of rat epididymal basal cells using a novel in vitro organoid modeldagger. Biol. Reprod. 2021, 105, 987–1001. [Google Scholar] [CrossRef]
  85. Seaberg, R.M.; van der Kooy, D. Stem and progenitor cells: The premature desertion of rigorous definitions. Trends Neurosci. 2003, 26, 125–131. [Google Scholar] [CrossRef]
  86. Rinaldi, V.D.; Donnard, E.; Gellatly, K.; Rasmussen, M.; Kucukural, A.; Yukselen, O.; Garber, M.; Sharma, U.; Rando, O.J. An atlas of cell types in the mouse epididymis and vas deferens. Elife 2020, 9, e55474. [Google Scholar] [CrossRef] [PubMed]
  87. Abe, K.; Takano, H.; Ito, T. Ultrastructure of the mouse epididymal duct with special reference to the regional differences of the principal cells. Arch. Histol. Jpn. 1983, 46, 51–68. [Google Scholar] [CrossRef]
  88. Dufresne, J.; Finnson, K.W.; Gregory, M.; Cyr, D.G. Expression of multiple connexins in the rat epididymis indicates a complex regulation of gap junctional communication. Am. J. Physiol. Cell Physiol. 2003, 284, C33–C43. [Google Scholar] [CrossRef]
  89. Tadokoro, T.; Wang, Y.; Barak, L.S.; Bai, Y.; Randell, S.H.; Hogan, B.L. IL-6/STAT3 promotes regeneration of airway ciliated cells from basal stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, E3641–E3649. [Google Scholar] [CrossRef]
  90. Yang, Y.; Riccio, P.; Schotsaert, M.; Mori, M.; Lu, J.; Lee, D.K.; Garcia-Sastre, A.; Xu, J.; Cardoso, W.V. Spatial-Temporal Lineage Restrictions of Embryonic p63(+) Progenitors Establish Distinct Stem Cell Pools in Adult Airways. Dev. Cell 2018, 44, 752–761.e4. [Google Scholar] [CrossRef]
  91. Clermont, Y.; Flannery, J. Mitotic activity in the epithelium of the epididymis in young and old adult rats. Biol. Reprod. 1970, 3, 283–292. [Google Scholar] [CrossRef] [PubMed]
  92. Cordero-Espinoza, L.; Dowbaj, A.M.; Kohler, T.N.; Strauss, B.; Sarlidou, O.; Belenguer, G.; Pacini, C.; Martins, N.P.; Dobie, R.; Wilson-Kanamori, J.R.; et al. Dynamic cell contacts between periportal mesenchyme and ductal epithelium act as a rheostat for liver cell proliferation. Cell Stem Cell 2021, 28, 1907–1921.e8. [Google Scholar] [CrossRef]
  93. Pratama, G.; Vaghjiani, V.; Tee, J.Y.; Liu, Y.H.; Chan, J.; Tan, C.; Murthi, P.; Gargett, C.; Manuelpillai, U. Changes in culture expanded human amniotic epithelial cells: Implications for potential therapeutic applications. PLoS ONE 2011, 6, e26136. [Google Scholar] [CrossRef] [PubMed]
  94. Chen, Z.H.; Luo, X.C.; Yu, C.R.; Huang, L. Matrix metalloprotease-mediated cleavage of neural glial-related cell adhesion molecules activates quiescent olfactory stem cells via EGFR. Mol. Cell Neurosci. 2020, 108, 103552. [Google Scholar] [CrossRef] [PubMed]
  95. Serrano Martinez, P.; Maimets, M.; Bron, R.; van Os, R.; de Haan, G.; Pringle, S.; Coppes, R.P. Role of quiescent cells in the homeostatic maintenance of the adult submandibular salivary gland. iScience 2022, 25, 105047. [Google Scholar] [CrossRef] [PubMed]
  96. Cao, W.; Chen, K.; Bolkestein, M.; Yin, Y.; Verstegen, M.M.A.; Bijvelds, M.J.C.; Wang, W.; Tuysuz, N.; Ten Berge, D.; Sprengers, D.; et al. Dynamics of Proliferative and Quiescent Stem Cells in Liver Homeostasis and Injury. Gastroenterology 2017, 153, 1133–1147. [Google Scholar] [CrossRef] [PubMed]
  97. Basak, O.; Beumer, J.; Wiebrands, K.; Seno, H.; van Oudenaarden, A.; Clevers, H. Induced Quiescence of Lgr5+ Stem Cells in Intestinal Organoids Enables Differentiation of Hormone-Producing Enteroendocrine Cells. Cell Stem Cell 2017, 20, 177–190.e4. [Google Scholar] [CrossRef] [PubMed]
  98. Vennekens, A.; Laporte, E.; Hermans, F.; Cox, B.; Modave, E.; Janiszewski, A.; Nys, C.; Kobayashi, H.; Malengier-Devlies, B.; Chappell, J.; et al. Interleukin-6 is an activator of pituitary stem cells upon local damage, a competence quenched in the aging gland. Proc. Natl. Acad. Sci. USA 2021, 118, e2100052118. [Google Scholar] [CrossRef] [PubMed]
  99. Mou, H.; Vinarsky, V.; Tata, P.R.; Brazauskas, K.; Choi, S.H.; Crooke, A.K.; Zhang, B.; Solomon, G.M.; Turner, B.; Bihler, H.; et al. Dual SMAD Signaling Inhibition Enables Long-Term Expansion of Diverse Epithelial Basal Cells. Cell Stem Cell 2016, 19, 217–231. [Google Scholar] [CrossRef]
  100. Edmondson, R.; Broglie, J.J.; Adcock, A.F.; Yang, L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay. Drug Dev. Technol. 2014, 12, 207–218. [Google Scholar] [CrossRef] [PubMed]
  101. Gu, L.; Mooney, D.J. Biomaterials and emerging anticancer therapeutics: Engineering the microenvironment. Nat. Rev. Cancer 2016, 16, 56–66. [Google Scholar] [CrossRef]
  102. Huh, D.; Hamilton, G.A.; Ingber, D.E. From 3D cell culture to organs-on-chips. Trends Cell Biol. 2011, 21, 745–754. [Google Scholar] [CrossRef]
  103. Smith, E.; Cochrane, W.J. Cystic Organoid Teratoma: (Report of a Case). Can. Med. Assoc. J. 1946, 55, 151–152. [Google Scholar] [PubMed]
  104. Lancaster, M.A.; Knoblich, J.A. Organogenesis in a dish: Modeling development and disease using organoid technologies. Science 2014, 345, 1247125. [Google Scholar] [CrossRef] [PubMed]
  105. Barker, N.; van Es, J.H.; Kuipers, J.; Kujala, P.; van den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007, 449, 1003–1007. [Google Scholar] [CrossRef] [PubMed]
  106. Sato, T.; Vries, R.G.; Snippert, H.J.; van de Wetering, M.; Barker, N.; Stange, D.E.; van Es, J.H.; Abo, A.; Kujala, P.; Peters, P.J.; et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 2009, 459, 262–265. [Google Scholar] [CrossRef] [PubMed]
  107. Ootani, A.; Li, X.; Sangiorgi, E.; Ho, Q.T.; Ueno, H.; Toda, S.; Sugihara, H.; Fujimoto, K.; Weissman, I.L.; Capecchi, M.R.; et al. Sustained in vitro intestinal epithelial culture within a Wnt-dependent stem cell niche. Nat. Med. 2009, 15, 701–706. [Google Scholar] [CrossRef] [PubMed]
  108. Busslinger, G.A.; Weusten, B.L.A.; Bogte, A.; Begthel, H.; Brosens, L.A.A.; Clevers, H. Human gastrointestinal epithelia of the esophagus, stomach, and duodenum resolved at single-cell resolution. Cell Rep. 2021, 34, 108819. [Google Scholar] [CrossRef] [PubMed]
  109. Broutier, L.; Mastrogiovanni, G.; Verstegen, M.M.; Francies, H.E.; Gavarro, L.M.; Bradshaw, C.R.; Allen, G.E.; Arnes-Benito, R.; Sidorova, O.; Gaspersz, M.P.; et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 2017, 23, 1424–1435. [Google Scholar] [CrossRef] [PubMed]
  110. Wang, S.; Wang, X.; Tan, Z.; Su, Y.; Liu, J.; Chang, M.; Yan, F.; Chen, J.; Chen, T.; Li, C.; et al. Human ESC-derived expandable hepatic organoids enable therapeutic liver repopulation and pathophysiological modeling of alcoholic liver injury. Cell Res. 2019, 29, 1009–1026. [Google Scholar] [CrossRef]
  111. Cerrizuela, S.; Kaya, O.; Kremer, L.P.M.; Sarvari, A.; Ellinger, T.; Straub, J.; Brunken, J.; Sanz-Morejón, A.; Korkmaz, A.; Martín-Villalba, A. High-throughput scNMT protocol for multiomics profiling of single cell s from mouse brain and pancreatic organoids. STAR Protoc. 2022, 3, 101555. [Google Scholar] [CrossRef]
  112. Drost, J.; Karthaus, W.R.; Gao, D.; Driehuis, E.; Sawyers, C.L.; Chen, Y.; Clevers, H. Organoid culture systems for prostate epithelial and cancer tissue. Nat. Protoc. 2016, 11, 347–358. [Google Scholar] [CrossRef]
  113. Ranjan, M.; Lee, O.; Cottone, G.; Mirzaei Mehrabad, E.; Spike, B.T.; Zeng, Z.; Yadav, S.; Chatterton, R.; Kim, J.J.; Clare, S.E.; et al. Progesterone receptor antagonists reverse stem cell expansion and the paracrine effectors of progesterone action in the mouse mammary gland. Breast Cancer Res. 2021, 23, 78. [Google Scholar] [CrossRef] [PubMed]
  114. Yang, Y.; Huang, R.; Cao, Z.; Ma, S.; Chen, D.; Wang, Z.; Feng, Y.; Lei, Y.; Zhang, Q.; Huang, Y. In vitro reconstitution of the hormone-responsive testicular organoids from murine primary testicular cells. Biofabrication 2022, 15, 015001. [Google Scholar] [CrossRef] [PubMed]
  115. Alves-Lopes, J.P.; Stukenborg, J.B. Testicular organoids: A new model to study the testicular microenvironment in vitro? Hum. Reprod. Update 2018, 24, 176–191. [Google Scholar] [CrossRef] [PubMed]
  116. Richer, G.; Baert, Y.; Goossens, E. In-vitro spermatogenesis through testis modelling: Toward the generation of testicular organoids. Andrology 2020, 8, 879–891. [Google Scholar] [CrossRef] [PubMed]
  117. Turco, M.Y.; Gardner, L.; Hughes, J.; Cindrova-Davies, T.; Gomez, M.J.; Farrell, L.; Hollinshead, M.; Marsh, S.G.E.; Brosens, J.J.; Critchley, H.O.; et al. Long-term, hormone-responsive organoid cultures of human endometrium in a chemically defined medium. Nat. Cell Biol. 2017, 19, 568–577. [Google Scholar] [CrossRef] [PubMed]
  118. Chumduri, C.; Turco, M.Y. Organoids of the female reproductive tract. J. Mol. Med. 2021, 99, 531–553. [Google Scholar] [CrossRef] [PubMed]
  119. Lin, Y.; Wei, Y.; Jiang, M.; Tang, X.; Huang, F.; Yang, X. Organoid culture of mouse fallopian tube epithelial stem cells with a thermo-reversible gelation polymer. Tissue Cell 2021, 73, 101622. [Google Scholar] [CrossRef]
  120. Li, X.Y.; Zheng, M.; Xu, B.; Li, D.L.; Shen, Y.; Nie, Y.Q.; Ma, L.; Wu, J. Generation of offspring-producing 3D ovarian organoids derived from female germline stem cells and their application in toxicological detection. Biomaterials 2021, 279, 121213. [Google Scholar] [CrossRef]
  121. Leir, S.H.; Yin, S.; Kerschner, J.L.; Xia, S.; Ahmadi, S.; Bear, C.; Harris, A. An organoid model to assay the role of CFTR in the human epididymis epithelium. Cell Tissue Res. 2020, 381, 327–336. [Google Scholar] [CrossRef]
  122. Cyr, D.G.; Pinel, L. Emerging organoid models to study the epididymis in male reproductive toxicology. Reprod. Toxicol. 2022, 112, 88–99. [Google Scholar] [CrossRef]
  123. Dufresne, J.; Gregory, M.; Pinel, L.; Cyr, D.G. Differential gene expression and hallmarks of stemness in epithelial cells of the developing rat epididymis. Cell Tissue Res. 2022, 389, 327–349. [Google Scholar] [CrossRef]
  124. Lu, C.; Le, Q. Advances in Organoid Technology: A Focus on Corneal Limbal Organoids. Stem Cell Rev. Rep. 2024, 20, 1227–1235. [Google Scholar] [CrossRef]
  125. Soto-Gamez, A.; Gunawan, J.P.; Barazzuol, L.; Pringle, S.; Coppes, R.P. Organoid-based personalized medicine: From tumor outcome prediction to autologous transplantation. Stem Cells 2024, 42, 499–508. [Google Scholar] [CrossRef] [PubMed]
  126. Moore, H.D.; Hartman, T.D. In-vitro development of the fertilizing ability of hamster epididymal spermatozoa after co-culture with epithelium from the proximal cauda epididymidis. J. Reprod. Fertil. 1986, 78, 347–352. [Google Scholar] [CrossRef] [PubMed]
  127. Patricio, D.; Santiago, J.; Mano, J.F.; Fardilha, M. Organoids of the male reproductive system: Challenges, opportunities, and their potential use in fertility research. WIREs Mech. Dis. 2023, 15, e1590. [Google Scholar] [CrossRef] [PubMed]
  128. Kristensen, D.M.; Nielsen, J.E.; Kalisz, M.; Dalgaard, M.D.; Audouze, K.; Larsen, M.E.; Jacobsen, G.K.; Horn, T.; Brunak, S.; Skakkebaek, N.E.; et al. OCT4 and downstream factors are expressed in human somatic urogenital epithelia and in culture of epididymal spheres. Mol. Hum. Reprod. 2010, 16, 835–845. [Google Scholar] [CrossRef]
  129. Kanbar, M.; Vermeulen, M.; Wyns, C. Organoids as tools to investigate the molecular mechanisms of male infertility and its treatments. Reproduction 2021, 161, R103–R112. [Google Scholar] [CrossRef]
  130. Sakib, S.; Yu, Y.; Voigt, A.; Ungrin, M.; Dobrinski, I. Generation of Porcine Testicular Organoids with Testis Specific Architecture using Microwell Culture. J. Vis. Exp. 2019, 152, e60387. [Google Scholar]
  131. Cala, G.; Sina, B.; De Coppi, P.; Giobbe, G.G.; Gerli, M.F.M. Primary human organoids models: Current progress and key milestones. Front. Bioeng. Biotechnol. 2023, 11, 1058970. [Google Scholar] [CrossRef]
  132. Li, H.; Gao, L.; Ye, Z.; Du, J.; Li, W.; Liang, L.; Zeng, Q.; Xi, J.; Yue, W.; Li, Z. Protective effects of resveratrol on the ethanol-induced disruption of retinogenesis in pluripotent stem cell-derived organoids. FEBS Open Bio 2023, 13, 845–866. [Google Scholar] [CrossRef]
  133. Sun, X.C.; Kong, D.F.; Zhao, J.; Faber, K.N.; Xia, Q.; He, K. Liver organoids: Established tools for disease modeling and drug development. Hepatol. Commun. 2023, 7, e0105. [Google Scholar] [CrossRef] [PubMed]
  134. Yoon, J.C.; Casella, J.L.; Litvin, M.; Dobs, A.S. Male reproductive health in cystic fibrosis. J. Cyst. Fibros. 2019, 18 (Suppl. 2), S105–S110. [Google Scholar] [CrossRef] [PubMed]
  135. Dai, X.; Gan, W.; Li, X.; Wang, S.; Zhang, W.; Huang, L.; Liu, S.; Zhong, Q.; Guo, J.; Zhang, J.; et al. Prostate cancer-associated SPOP mutations confer resistance to BET inhibitors through stabilization of BRD4. Nat. Med. 2017, 23, 1063–1071. [Google Scholar] [CrossRef] [PubMed]
  136. Yan, Y.; Ma, J.; Wang, D.; Lin, D.; Pang, X.; Wang, S.; Zhao, Y.; Shi, L.; Xue, H.; Pan, Y.; et al. The novel BET-CBP/p300 dual inhibitor NEO2734 is active in SPOP mutant and wild-type prostate cancer. EMBO Mol. Med. 2019, 11, e10659. [Google Scholar] [CrossRef] [PubMed]
  137. Sharma, I.; Kumari, P.; Sharma, A.; Saha, S.C. SARS-CoV-2 and the reproductive system: Known and the unknown!! Middle East. Fertil. Soc. J. 2021, 26, 1. [Google Scholar] [CrossRef] [PubMed]
  138. Tian, Y.; Zhou, L.Q. Evaluating the impact of COVID-19 on male reproduction. Reproduction 2021, 161, R37–R44. [Google Scholar] [CrossRef] [PubMed]
  139. Zuchowska, A.; Baranowska, P.; Flont, M.; Brzozka, Z.; Jastrzebska, E. Review: 3D cell models for organ-on-a-chip applications. Anal. Chim. Acta 2024, 1301, 342413. [Google Scholar] [CrossRef] [PubMed]
  140. Moore, H.D.; Akhondi, M.A. In vitro maturation of mammalian spermatozoa. Rev. Reprod. 1996, 1, 54–60. [Google Scholar] [CrossRef] [PubMed]
  141. Kervancioglu, M.E.; Saridogan, E.; Aitken, R.J.; Djahanbakhch, O. Importance of sperm-to-epithelial cell contact for the capacitation of human spermatozoa in fallopian tube epithelial cell cocultures. Fertil. Steril. 2000, 74, 780–784. [Google Scholar] [CrossRef] [PubMed]
  142. Partiot, E.; Hirschler, A.; Colomb, S.; Lutz, W.; Claeys, T.; Delalande, F.; Deffieu, M.S.; Bare, Y.; Roels, J.R.E.; Gorda, B.; et al. Brain exposure to SARS-CoV-2 virions perturbs synaptic homeostasis. Nat. Microbiol. 2024, 9, 1189–1206. [Google Scholar] [CrossRef]
Figure 1. Schematic diagram of the cellular organization in a representative cross-section of the epididymis. Modified and reprinted with permission of the author (Chen et al., 2022) [24].
Figure 1. Schematic diagram of the cellular organization in a representative cross-section of the epididymis. Modified and reprinted with permission of the author (Chen et al., 2022) [24].
Biomolecules 14 01026 g001
Figure 2. Timeline for the development of epididymal organoid cultures. A summary of key landmark studies and breakthroughs leading to the establishment of epididymal organoids in different species including human, mouse, and rat. Two-dimensional, (2D); three-dimensional, (3D).
Figure 2. Timeline for the development of epididymal organoid cultures. A summary of key landmark studies and breakthroughs leading to the establishment of epididymal organoids in different species including human, mouse, and rat. Two-dimensional, (2D); three-dimensional, (3D).
Biomolecules 14 01026 g002
Figure 3. Simplified workflow for organoid formation from mouse epididymis epithelial cells in our laboratory. Epididymis from an adult mouse was sampled and enzymatically digested to obtain single-cell suspensions. The cells were cultured in extracellular matrix (basement membrane extract—BME) under 3D conditions and subsequently incubated at a temperature of 34 °C.
Figure 3. Simplified workflow for organoid formation from mouse epididymis epithelial cells in our laboratory. Epididymis from an adult mouse was sampled and enzymatically digested to obtain single-cell suspensions. The cells were cultured in extracellular matrix (basement membrane extract—BME) under 3D conditions and subsequently incubated at a temperature of 34 °C.
Biomolecules 14 01026 g003
Table 1. Characteristics, functions, and markers of the epididymal epithelial cell.
Table 1. Characteristics, functions, and markers of the epididymal epithelial cell.
Cell TypeCharacteristicFunctionMarkerReferences
Principal cellTall, columnar shape in the proximal regions with a squared-off appearance in the distal regions, with microvilli 500 nm–1.0 mm in length and 100 nm in width forming the epididymal brush borderSecretion/Reabsorb, Merocrine, Apocrine secretionsAQP-9, CFTR, NHER1[11,25,26]
Clear cellAn apical pole enriched with mitochondria which displays a complete and functional endocytic apparatusEndocytic cells, proton secretionV-ATPase, CIC-5[27,28]
Apical cellPresent in the initial segment of the epididymis displaying a spherical nucleus at the apical pole of the epitheliumControl of inflammatory responses in the epididymisV-ATPase, GSTM3[11,29,30]
Basal cellPyramidal-shaped cells located at the base of the epithelium which directly interact with neighboring principal and clear cells through gap junctions“Stem cell” character and “lumen-reaching” propertyKRT5[11,23]
Narrow cellElongated and narrow shape, present in the initial segment of the epididymisProton secretion and acidification of the epididymal fluidV-ATPase, CIC-5[27,28]
Table 2. Timeline for the development of epididymal organoids in different species.
Table 2. Timeline for the development of epididymal organoids in different species.
YearSpeciesResultsReferences
1992HumanEpididymal fragments formed everted epithelial spheres that maintained cell integrity for 5–7 days.[34]
2010HumanEpididymal cells formed spheres for at least 20 days.[128]
2015RatBasal cells may represent an epididymal stem cell population.[13]
2016MouseExpanded epididymis basal cells efficiently generated organoids in Matrigel.[99]
2020HumanEpididymal cells generated organoid and provided the tool for studying cystic fibrosis (CF) in infertile men.[121]
2021–2022RatBasal cells generated organoids capable of secreting function and columnar cells represent an epididymal stem/progenitor cell population.[84,123]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Nie, J.; Chen, H.; Zhao, X. Advancement and Potential Applications of Epididymal Organoids. Biomolecules 2024, 14, 1026. https://doi.org/10.3390/biom14081026

AMA Style

Nie J, Chen H, Zhao X. Advancement and Potential Applications of Epididymal Organoids. Biomolecules. 2024; 14(8):1026. https://doi.org/10.3390/biom14081026

Chicago/Turabian Style

Nie, Junyu, Hao Chen, and Xiuling Zhao. 2024. "Advancement and Potential Applications of Epididymal Organoids" Biomolecules 14, no. 8: 1026. https://doi.org/10.3390/biom14081026

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop