Skip to main content

Expression of NELL2/NICOL-ROS1 lumicrine signaling-related molecules in the human male reproductive tract

Abstract

The maturation of spermatozoa is a regulated process, influenced by genes expressing essential secreted proteins in the proximal epididymis. Recent genetic studies in rodents have identified the non-sex steroidal molecular signals that regulate gene expression in the proximal epididymis. Germ cells in the testis secrete ligand proteins into the seminiferous tubule lumen The ligand proteins travel through the male reproductive tract lumen to the epididymis, where they bind to receptors, triggering the differentiation of the luminal epithelium for sperm maturation. It is, however, not fully unveiled if such a testis-epididymis trans-luminal signaling mechanism exists in other species, especially humans. In the present study, the rodent-type testis-epididymis trans-luminal signaling in the human male reproductive tract was evaluated in a step-by-step manner by analyzing testis and epididymis gene expression and signaling mediator protein function. There was a significant correlation between the epididymal expressions of mouse genes upregulated by the trans-luminal signaling and those of their human orthologs, as evaluated by the correlation coefficient of 0.604. The transcript expression of NELL2 and NICOL encoding putative ligand proteins was also observed in human testicular cells. In vitro experiments demonstrated that purified recombinant human NELL2 and NICOL formed a molecular complex with similar properties to rodent proteins, which was evaluated by a dissociation equilibrium constant of 110 nM. Recombinant human NELL2 also specifically bound to its putative receptor human ROS1 in vitro. Collectively, these findings suggest that the rodent-type testis-epididymis secreted signaling mechanism is also possible in the human male reproductive tract.

Introduction

Production of competent gametes is the prerequisite for successful reproduction. In aquatic vertebrates, spermatozoa generated in the testes are fully capable of fertilization. In terrestrial vertebrates such as mammals, spermatozoa generated in the testicular seminiferous tubules are still immature and need further functional maturation to acquire full fertilizing ability. The maturation of spermatozoa occurs at the post-testicular level; the testicular spermatozoa are transported via the efferent duct to the epididymis, where they become functionally mature. The epididymis is a highly coiled epithelial duct constituting a part of the sperm transport route. The spermatozoa develop the fertilizing ability as they pass through the lumen of the epididymis. In the epididymis, various proteins including secreted protein families such as β-defensins, lipocalins, and cystatin-related epididymal spermatogenic (CRES) proteins are specifically expressed to modify the fertilizing ability of spermatozoa [1,2,3].

The mechanisms of sperm maturation in the epididymis are orchestrated by the testes. Endocrine control of epididymal gene expression and function is facilitated by sex steroids such as androgens and estrogens synthesized by Leydig cells within the testes. These sex steroids are transported through the bloodstream to the epididymis, where they interact with their receptors, thereby modulating gene expressions and cellular functions, as shown by gene targeting strategies [4, 5]. In addition to the endocrine action of sex steroids, a mechanism involving the transmission of signals by non-steroidal factors through the reproductive tract has been identified in rodents. When solely ligating the efferent duct while leaving the testes intact, the differentiation of the luminal epithelial cells of the initial segment (IS) epididymis and subsequent induction of gene expression becomes disrupted [6, 7]. This experimental evidence strongly indicates the presence of a signaling mechanism, through which the testes regulate cellular processes in the epididymis via the efferent duct, at least in rodents. Since this type of secretion signaling between the testes and epididymis acts through the lumen, it has been referred to as “lumicrine,” a terminology introduced by Barry T. Hinton [8].

Utilizing reverse genetic analyses in genetically modified mice, key components involved in this testis-epididymis signaling network have been identified. Neural EGF-like like 2 (NELL2) and the NELL2-interacting cofactor for lumicrine signaling (NICOL) serve as ligand proteins secreted by the germ cells within the testes into the reproductive tract lumen [9, 10]. These ligand proteins traverse the reproductive tract lumen to reach the epididymis, where they bind to the receptor-type tyrosine kinase ROS1 expressed in the IS [11,12,13]. Upon ligand binding, ROS1 activates downstream intracellular signaling pathways [14, 15]. These pathways lead to the induction of luminal epithelial cell differentiation in the IS region and the expression of various genes including β-defensins, lipocalins, CRES/cystatins, proteases, and other secreted proteins essential for sperm maturation [1,2,3, 9]. The testis-epididymal trans-luminal signaling in the male reproductive tract is thus apparent in rodents as schematically summarized in Fig. 1.

Fig. 1
figure 1

A schematic representation of testis-epididymis trans-luminal signaling in rodents. (A) A schematic representation of rodent male reproductive tract. (B) A schematic representation of testis-epididymis trans-luminal signaling lumicrine. In the testicular seminiferous tubule, lumicrine factors such as NELL2 and NICOL are secreted from germ cells located inside the blood-testis barrier. The secreted ligand proteins go trans-luminally through the efferent duct to the epididymis. In the epididymis, the testis-derived ligands bind to the receptor ROS1 on the apical surface of the epididymal luminal epithelium. Upon ROS1 activation, genes necessary for epididymal differentiation and sperm maturation are induced

The tall pseudostratified luminal epithelium of the IS triggered to differentiate by the trans-luminal secreted signaling is a prominent histological feature characteristic of rodents [6, 7, 9, 10, 14]. The testis-epididymis trans-luminal signaling system and its regulation of epididymal functions in humans have not been fully investigated, because the human epididymis lacks anatomical features such as the prominent epididymal IS in rodents [16, 17], implying another mechanism distinct from murine lumicrine signaling. However, the tall luminal epithelium of the rodent epididymal IS has not been shown to be the universal indicator of lumicrine-regulated epididymal function including sperm maturation across species although it is a good indicator of lumicrine signaling in rodents. Therefore, it is necessary to evaluate carefully the possibility of testis-epididymis trans-luminal signaling in humans based also on non-histological data.

The bioinformatic approaches using next-generation sequencing data have significantly enhanced the capabilities of gene expression analyzes and supplement histological approaches. By conducting genome-wide analyses, in addition to individual gene expression, the regulatory mechanisms working behind the specific biological processes can be investigated. In addition to such gene expression analysis, protein function analyzes are also valuable in characterizing biological processes. In the present study, the potential testis-epididymis communication through the male reproductive tract in humans was evaluated by transcriptome and protein biochemical analyzes.

Materials and methods

Epididymal anatomy

In the mouse transcriptome analyses, the IS and caput were combined, and such a tissue dissection was indicated by the description “IS-caput” throughout the manuscript.

Transcriptome analyses

The following human and transcriptome datasets publicly available were downloaded from the NCBI Gene Expression Omnibus website (https://www.ncbi.nlm.nih.gov/geo/): GSE150852, RNA-seq data of human epididymis [18]; GSE133920, RNA-seq data of WT and Nell2 knockout (KO) mouse caput epididymis [9]. To convert gene accession numbers into gene names, the DAVID Bioinformatics Resources website (https://david.ncifcrf.gov/home.jsp) was used. The transcriptome data were incorporated into Microsoft Excel software for further analysis. The human testis single-cell RNA-seq dataset, which contains information about gene expression in individual cells and cell population clustering, was downloaded from the Mendeley Data website (https://data.mendeley.com/datasets/kxd5f8vpt4/1) [19]. The dataset was imported into Loupe Browser software 6.1.0 (10x Genomics, Inc.) to extract the expression profiles for human NELL2 and NICOL transcripts. Human orthologs of mouse genes were identified on the NCBI Gene website (https://www.ncbi.nlm.nih.gov/gene/).

Plasmid construction

cDNAs encoding C-terminally 6×His-tagged, human NELL2, C-terminally 6×His-tagged human NICOL, and C-terminally 8xHis and Rho1D4-tagged human ROS1 ectodomain were cloned into a pCAG vector containing the CAG promoter and a rabbit globin poly(A) signal. A C-terminally 6×His-tagged cartilage oligomeric matrix protein (COMP) expression plasmid was constructed previously [9].

Protein purification

Human 293-F cells (Invitrogen, #R79007), a derivative of HEK293T cells, was used for recombinant protein expression. 293-F cells were cultured in suspension in HE400/AZ chemically defined medium (Gmep Inc., Japan, #HE400AZ-0010). A C-terminally 6×His-tagged COMP was expressed in 293-F cells and purified from the conditioned medium as described previously [9]. A C-terminally 6×His-tagged recombinant human NELL2 protein and C-terminally 6×His-tagged human NICOL protein were transiently expressed in 293-F cells by transfecting expression plasmids using a Gxpress 293 Transfection Kit (Gmep Inc., #GX293-RK-0010) according to manufacturer’s instructions. After 3 to 5 days of culture, the conditioned medium was harvested by brief centrifugation and the proteins were precipitated by ammonium sulfate at a final concentration of 80%. The precipitate isolated by filtration was then dissolved in 20 mM Tris-HCl pH 8.0, 30 mM imidazole, and 1 mM phenylmethylsulfonyl fluoride and loaded onto nickel affinity resin (Qiagen, #30,210). After washing with 20 mM Tris-HCl pH 8.0, 30 mM imidazole, and 150 mM NaCl, the bound protein was eluted with 300 mM imidazole pH 8.0, desalted using a PD-10 gel filtration column (Cytiva, #17,085,101) pre-equilibrated with phosphate-buffered saline (PBS). Recombinant 6×His-tagged human NELL2 and 6×His-tagged human NICOL were validated by SDS-PAGE under reducing conditions and subsequent immunoblot detection with anti-NELL2 antibody and anti-NICOL antibody, respectively. The concentrations of purified recombinant proteins were determined with the Pierce™ BCA Protein Assay Kit (Thermo Fisher Scientific Inc., #23,225) relative to bovine serum albumin (BSA) as the standard. The absorbance at 560 nm was monitored using an iMark microplate reader (Bio-Rad Laboratories, Inc.). Purified proteins were stored at − 80 °C until use.

Immunoblot analyses

Proteins were separated by SDS-PAGE under reducing conditions, and blot onto polyvinylidene difluoride membrane using Trans-blot turbo system (Bio-rad, #1704150J1). The membranes were incubated with a blocking solution (3% BSA and 0.05% Tween20 in Tris-buffered saline) for 30 min, then with primary antibodies diluted in the blocking solution overnight. The bound antibodies were detected by following incubation with peroxidase-conjugated secondary antibodies and chemiluminescent reaction using Chemi-Lumi One Super (Nacalai Tesque, #02230). The chemiluminescent signals were detected using Amersham ImageQuant 800 (Cytiva). Antibodies used were as follows: rabbit polyclonal anti-NELL2 antibody (Proteintech, #11268-1-P), rabbit polyclonal anti-NICOL antibody (Atlas antibodies, #HPA052447), mouse monoclonal anti-Rho1D4 antibody (Cube biotech, # 40,020), peroxidase-conjugated goat polyclonal anti-rabbit IgG (Jackson Immunoresearch, #111-036-045), and perpoxydase-conjugated goat polyclonal anti-mouse IgG (#115-036-062, Jackson Immunoresearch). Uncropped immunoblot images were provided as Supplementary Material 1.

Protein interaction analyses

To investigate protein interactions with human ROS1, a total of 100 µg of each purified protein was conjugated with a 25-µL bed volume of NHS-activated agarose (Thermo Fisher Scientific Inc., #26,200). For NELL2 pulldown, purified recombinant NELL2 was applied to protein-conjugated beads, and for ROS1 pulldown, ROS1 ectodomain tagged with 8xHis and Rho1D4 was transiently expressed in 293 F cells, and the transfected cells were lysed with lysis buffer. Cell lysate (1 mL of 1 mg protein/mL) was mixed with a 25-µL bed volume of protein-conjugated agarose beads and the mixture was incubated overnight at 4 °C with gentle rotation, washed three times with 1 mL of lysis buffer, and subjected to SDS-PAGE to separate the bound proteins for subsequent immunoblot analysis. Anti-NELL2 antibody and anti-Rho1D4 antibody were used to detect NELL2 and ROS1 ectodomain, respectively.

To determine kinetic parameters, a surface plasmon resonance assay was carried out by immobilizing purified human NELL2 protein onto a series S sensor chip CM5 (Cytiva, #29,104,988) as a ligand. Purified human NICOL protein dissolved in PBS was loaded as an analyte and its association and dissociation kinetics were monitored using a Biacore T200 instrument (Cytiva). The association/dissociation dynamics of NICOL to NELL2 at increasing concentration were monitored to determine the association rate constant ka, dissociation rate constant kd, and dissociation equilibrium constant KD.

Statistical analysis

All experiments other than those using publicly available transcriptome data were repeated biologically at least thrice, and similar results were obtained. Student’s t-test p values and correlation coefficient r values were calculated with Microsoft Excel 2019 (Microsoft Corporation).

Ethical approval

Not applicable.

Drawings

Schematic drawings were generated using Microsoft PowerPoint software.

Data availability

Transcriptome data supporting the results of this study are available at the NCBI GEO under accession numbers GSE150852 and GSE133920. The single-cell RNA-seq data supporting the results of this study are available at the Mendeley Data website (URL: https://data.mendeley.com/datasets/kxd5f8vpt4/1).

Results

Bioinformatic characterization of human epididymis gene expression

In the murine epididymal IS, many genes are regulated under the control of testis-epididymis trans-luminal signaling; such genes are downregulated in the epididymal IS of Nell2 KO, Nicol KO, Ros1 KO, W/Wv, or efferent duct-ligated mice in which testis-epididymis trans-luminal signaling is genetically or experimentally ablated [9, 10, 20]. To characterize gene expression in the human epididymis based on findings in mice, human genes orthologous to mouse genes expressed in the IS-caput epididymis are analyzed.

Mouse genes expressed in the IS-caput epididymis were enriched in three different ways (I ~ III) and the human orthologs of such enriched mouse genes were identified (Table 1): (I) top 400 genes highly expressed in the WT mouse IS epididymis, 353 orthologs existed in humans, and the transcriptome data are available for 341 orthologs; (II) 450 genes relatively highly expressed in the WT mouse IS-caput epididymis (IS-caput/cauda > 100, t-test < 0.005), 372 orthologs existed in humans, and transcriptome data are available for 359 orthologs, (III) 342 genes significantly downregulated in Nell2 KO mouse IS-caput epididymis (Nell2 KO/WT < 0.3, t-test < 0.005), 295 orthologs existed in humans, and transcriptome data are available for 284 orthologs. The selected orthologs were represented in plots (Fig. 2a-c). The correlation coefficient r between mouse IS-caput epididymis and human caput epididymis for each group was calculated: r = 0.005, 0.078, and 0.604 for the criteria I, II, and III, respectively.

Fig. 2
figure 2

RNA-seq profiling of genes orthologous between human and mouse expressed in the caput epididymis. (A) Plot representation of orthologs abundantly expressed in the mouse IS-caput epididymis. (B) Plot representation of orthologs highly dominantly expressed in the mouse IS-caput epididymis (IS-caput/cauda > 10, t-test < 0.005). (C) Plot representation of orthologs whose expressions in the Nell2 KO IS-caput epididymis are downregulated (Nell2 KO/WT < 0.3, t-test < 0.005). The correlation coefficient r is indicated in each panel

Table 1 A Selection of mouse genes expressed in the epididymis and their human orthologs

Since the correlation constant r for selection criterion III is especially high, 19 genes, i.e., Lcn8/LCN8, Defb20/DEFB128, Lcn2/LCN2, Cst11/CST11, Defb18/DEFB113, Lcn10/LCN10, Crisp1/CRISP1, Ovch2/OVCH2, Adam28/ADAM28, Tmem150c/TMEM150C, Tpst2/TPST2, Rtcb/RTCB, Slc1a1/SLC1A1, Ppp1ca/PPP1CA, Dbi/DBI, Litaf/LITAF, Cldn10/CLDN10, Mfsd2a/MFSD2A, Dapl1/DAPL1, highly expressed in both human and mouse caput epididymides (mouse IS-caput > 20 RPKM and human caput > 20 reads per kilobase million [RPKM]) were picked out to interpret the result of selection based on the criterion III. The epididymal expression of these genes was visualized as follows: mouse IS-caput WT vs. Nell2 KO (Fig. 3a), mouse WT IS-caput, corpus, and cauda (Fig. 3b), human caput, corpus, and cauda (Fig. 3c). Most mouse genes were expressed higher in the IS-caput than in corpus and cauda epididymis, and not all but many human orthologs were also expressed higher in the caput than in the corpus and cauda epididymis.

Fig. 3
figure 3

Expressions of orthologs expressed in the mouse and human caput epididymis. (A) Mouse gene expression in the WT and Nell2 KO IS-caput epididymis. (B) Mouse gene expression in the WT IS-caput, corpus, and cauda epididymis. Note that Y axes are magnified for relatively low-expressing genes. (C) Human gene expression in the caput, corpus, and cauda epididymis Average ± SE is shown for 19 selected orthologs (mouse IS-caput > 20 RPKM and human caput > 20 RPKM) (left). A magnified view for relatively low-expressing genes is also shown (right)

Bioinformatic characterization of the testis-epididymis trans-luminal signaling in the human male reproductive tract

In the rodent male reproductive organs, NELL2 and NICOL are secreted in the seminiferous lumen and go from the testis through the lumen to the epididymis where they bind their receptor ROS1 to regulate IS differentiation and gene expression [9, 10]. In mice, Nell2 and Gm1673 encoding NELL2 and NICOL, respectively, are expressed in adluminally located cells such as the spermatocyte and spermatid subpopulations inside the seminiferous tubules and satisfy the criterion necessary for testis-epididymis trans-luminal signaling [9, 10]. To evaluate the possibility of testis-epididymis trans-luminal signaling in the human male reproductive tract, the expression of genes in the human male reproductive organs was bioinformatically explored by using single-cell RNA-seq data.

The human testis single-cell RNA-seq data [19] were analyzed to examine the expression patterns of NELL2 encoding NELL2 and C4orf48 (or NICOL1) encoding NICOL in the human testis. NELL2 was expressed in the cell populations of spermatogonia, preleptotene spermatocytes, leptotene/zygotene spermatocytes, and pachytene spermatocytes (Fig. 4A). C4orf48 was expressed in almost all cell populations (Fig. 4B). Therefore, NELL2 and C4orf48/NICOL1 were co-expressed in the cell populations of spermatogonia, preleptotene spermatocytes, leptotene/zygotene spermatocytes, and pachytene spermatocytes. Since spermatocytes at their later stages such as pachytene spermatocytes are located adluminally and their secreting proteins such as NELL2 and NICOL are included in the seminiferous fluid and able to go trans-luminally to the epididymis.

Fig. 4
figure 4

Expression profiles of NELL2 and NICOL encoding human orthologs of rodent testis-epididymis trans-luminal signaling ligands. (A,B) are t-distributed stochastic neighbour embedding (t-SNE) plots showing the expression of NELL2 (A) and NICOL (B) profiled by human testis single-cell RNA-seq

Characterization of protein interactions mediating testis-epididymis signaling

In the present study, molecular interaction assays using purified proteins were employed as much as possible to rigorously assess protein-protein interactions. In mice, NELL2 and NICOL constitute a ligand molecular complex [10]. The potential of interaction between human NELL2 and human NICOL was examined in vitro using recombinant proteins expressed in mammalian cells. C-terminally 6×His-tagged human NELL2 and NICOL proteins were transiently expressed in 293-F cells and purified from the conditioned media by nickel affinity chromatography (Fig. 5A-C).

Fig. 5
figure 5

In vitro characterization of molecular functions of human NELL2 and NICOL. (A) A schematic representation of recombinant protein expression and affinity purification. (B) Coomassie brilliant blue (CBB) staining of recombinant NELL2, recombinant COMP, BSA (left) and recombinant NICOL (right) proteins. (C) Immunoblot detection of recombinant human NELL2 (left) and human NICOL (right) with anti-NELL2 and anti-NICOL antibodies, respectively. (D) The association and dissociation of recombinant human NICOL bound to recombinant human NELL2 analyzed by surface plasmon resonance analysis. (E) Human ROS1 pulldown with human NELL2, COMP, and BSA. (F) Human ROS1 pulldown with human NICOL and BSA

The kinetic parameters of the interaction between human NELL2 and NICOL were determined by the surface plasmon resonance analysis using purified recombinant human NELL2 as a ligand and human NICOL as an analyte (Fig. 5D). The association rate constant ka and the dissociation rate constant kd were determined to be 9.1 × 103 M− 1s− 1 and 9.8 × 10− 4 s− 1, respectively. Consequently, the dissociation equilibrium constant KD = kd/ka was calculated to be 110 nM, which is comparable to the KD =80 nM for interaction between myc epitope and anti-myc monoclonal antibody 9E10 clone (KD =80 nM) [21], indicating the tight NELL2-NICOL interaction.

Using purified recombinant proteins, the interactions of NELL2 and NICOL with ROS1 were also investigated. As the 6×His-tagged recombinant human ROS1 protein was not able to be purified probably because of the steric hindrance to the C-terminally located 6×His tag by ROS1 protein itself, we used cell lysates of 293-F cells that were transiently expressing full-length human ROS1 for the interaction assay. When the cell lysates were mixed with recombinant human NELL2, COMP, or BSA, only human NELL2 specifically pulled down ROS1 (Fig. 5E). Human NICOL did not pull down ROS1 (Fig. 5F), indicating that the NICOL-ROS1 interaction is not strong enough to be detected by the assay employed. These findings elucidate unveiled the molecular capabilities of human NELL2 and NICOL to form a complex and interact with human ROS1, particularly through NELL2.

Discussion

In the mouse epididymis, the IS exhibits histologically prominent features characterized by pseudostratified luminal epithelium, making its regression easily observable through experimental interference of testis-epididymis trans-luminal signaling. Consequently, such histological features of the IS in mice have been employed as indicators of lumicrine control in rodents [6, 7, 9, 10, 14]. In contrast, the human caput epididymis does not exhibit pronounced luminal epithelium thickening as observed in the mouse IS epididymis [16, 17]. However, the absence of histological features akin to the mouse IS in the human caput epididymis does not provide evidence to conclude that the testis-epididymis trans-luminal signaling does not function in humans. In this study, we analyzed the possibility of testis-epididymis trans-luminal signaling in humans based on transcriptome and biochemical data.

Genes expressed in the human epididymis and whose orthologs are regulated by testis-epididymis trans-luminal signaling in mice

In the epididymis, various proteins including protein families such as β-defensins, lipocalins, and cystatin-related epididymal spermatogenic (CRES) proteins are abundantly expressed in the proximal epididymis including the IS and the caput, to modify the fertilizing ability of spermatozoa [22]. In the present study, genes characterizing mouse IS-caput were selected in three different ways: abundant expression, IS-caput-specific expression, and lumicrine-regulated expression. The correlation coefficient r = 0.604 between IS-caput expression of the lumicrine-regulated mouse genes and caput expression of their human ortholog is significantly high compared with the others, implying a possibility that lumicrine signaling may also be active in the human male reproductive tract.

Among the orthologs thus identified and abundantly expressed in the mouse and human proximal epididymis as shown in Fig. 3, included multiple genes essential for sperm maturation and male fertility. Mice lacking 8 cystatin genes (Cstl1, Cst11, Cstdc1, Cst12, Cst8, Cst13, Cst9, and Cstdc2) on chromosome 2 possess defective sperm maturation and male infertility [9, 23]. Lipocalins (Lcn8/LCN8, Lcn2/LCN2Lcn2, Lcn10/LCN10): mice lacking 5 lipocalin genes (Lcn5, Lcn6, Lcn8, Lcn9, and Lcn10) clustered on chromosome 2 possess abnormal sperm maturation and male subfertility [24]. Crisp1 KO male mice are fertile but possess reduced in vitro fertilizing ability [25]. Ovch2 and Adam28 encode secreted proteases. Mice lacking Ovch2 or Adam28 possess abnormal sperm maturation and complete infertility or reduced male fertility, respectively [9]. Tpst2, Altered sperm-egg interactions [26]. β-defensins constitute a large family which has evolved by gene duplication in each species and are expected to function in a compensatory and/or synergistic manner. There are no reports on whether Defb20 and Defb18 are essential for sperm maturation and male fertility, but deletion of nine β-defensin genes in the mouse results in defective sperm maturation and male sterility in mice [27] and the deletion of two or three β-defensin genes result in decreased sperm maturation in rats [28]. The facts support the possibility that there is a common regulatory mechanism between rodents and humans for gene expression and sperm maturation in the epididymis, although it should be confirmed experimentally further.

Testicular cells expressing testis-epididymis trans-luminal signaling ligands

A prerequisite to the trans-luminal action of testis-epididymis signaling is the expression of ligand proteins in the testicular cells facing the seminiferous lumen. Since spermatogonial stem cells, undifferentiated and differentiated spermatogonia, and preleptotene spermatocytes are all sequestered outside the blood-testis barrier (BTB) in the basal compartment, lumicrine factor expression should take place in cells such as late spermatocytes (e.g., zygotene, pachytene, and diplotene spermatocytes), secondary spermatocytes, spermatids, or spermatozoa restricted to the apical compartment in the seminiferous epithelium [29]. The observed NELL2 and NICOL expression in these cells satisfies the above criterion for testicular lumicrine factors. A preceding study demonstrating ROS1 expression in the human epididymis by in situ hybridization and qPCR analyses [30] suggests the potential of ROS1 to function as a lumicrine receptor in humans. The dissociation constant KD = 110 nM for human NELL2-human NICOL interaction determined by surface plasmon resonance analysis in the present study is comparable to the KD = 87 nM for mouse NELL2-mouse NICOL interaction [9], indicating that NELL2 and NICOL form a tight heteromeric complex also in humans. The ROS1 pulldown assay using purified NELL2 and NICOL detected NELL2-ROS1 interaction, indicating that a molecular complex by NELL2 and NICOL could be a ligand for ROS1. NICOL-ROS1 interaction should be further evaluated in both mice and humans to evaluate its role in lumicrine signaling. Collectively, these results suggest that the testicular NELL2 and NICOL and the epididymal ROS1 can constitute a lumicrine trans-luminal signaling system in the human male reproductive tract. An intriguing question is whether dysfunction of NELL2, NICOL, or ROS1 causes male infertility, although mutations in these genes have not been identified as causal factors in humans.

Evaluation of the whole processes of testis-epididymis trans-luminal signaling in the human male reproductive tract

The step-by-step processes of testis-epididymis trans-luminal signaling in the male reproductive tract of mice and humans and their supporting evidence were comparatively summarized in Table 2. Most cellular and molecular level events in testis-epididymis trans-luminal signaling were conserved between mice and humans, suggesting the possibility that trans-luminal signaling is also in humans. Whether lumicrine regulates gene expression in the proximal epididymis in humans is still unclear. However, it is worth noting that human orthologs of murine genes, which are regulated by lumicrine in the mouse epididymis, are also expressed in the human proximal epididymis.

Table 2 Summary of testis-epididymis trans-luminal signalling events in rodents and humans

It is still necessary to further examine new findings obtained from experimental animal studies with available human data on whether testis-epididymis trans-luminal signaling functions in humans and if so, to what extent it is the same and how it differs from that in experimental animals. As the volume of bioinformatic data has exploded in recent years, it is expected that the effective use of such data will lead to a further understanding of human testis-epididymis trans-luminal signaling.

Data availability

The data used and/or analysed during the current study are available from the corresponding author.

References

  1. Kirchhoff C, Habben I, Ivell R, Krull N. A major human epididymis-specific cDNA encodes a protein with sequence homology to extracellular proteinase inhibitors. Biol Reprod. 1991;45:350–7.

    Article  CAS  PubMed  Google Scholar 

  2. Kato M, Sung WK, Kato K, Goodman DS. Immunohistochemical studies on the localization of cellular retinol-binding protein in rat testis and epididymis. Biol Reprod. 1985;32:173–89.

    Article  CAS  PubMed  Google Scholar 

  3. Cornwall GA, Orgebin-Crist MC, Hann SR. The CRES gene: a unique testis-regulated gene related to the cystatin family is highly restricted in its expression to the proximal region of the mouse epididymis. Mol Endocrinol. 1992;6:1653–64.

    CAS  PubMed  Google Scholar 

  4. Krutskikh A, De Gendt K, Sharp V, Verhoeven G, Poutanen M, Huhtaniemi I. Targeted inactivation of the androgen receptor gene in murine proximal epididymis causes epithelial hypotrophy and obstructive azoospermia. Endocrinology. 2011;152:689–96.

    Article  CAS  PubMed  Google Scholar 

  5. Joseph A, Shur BD, Ko C, Chambon P, Hess RA. Epididymal hypo-osmolality induces abnormal sperm morphology and function in the estrogen receptor alpha knockout mouse. Biol Reprod. 2010;82:958–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Moniem KA, Glover TD, Lubicz-Nawrocki CW. Effects of duct ligation and orchidectomy on histochemical reactions in the hamster epididymis. J Reprod Fertil. 1978;54:173–6.

    Article  CAS  PubMed  Google Scholar 

  7. Fawcett DW, Hoffer AP. Failure of exogenous androgen to prevent regression of the initial segments of the rat epididymis after efferent duct ligation or orchidectomy. Biol Reprod [Internet]. 1979;20:162–81.

    Article  CAS  PubMed  Google Scholar 

  8. Hinton BT, Lan ZJ, Rudolph DB, Labus JC, Lye RJ. Testicular regulation of epididymal gene expression. J Reprod Fertil Suppl. 1998;53:47–57.

    CAS  PubMed  Google Scholar 

  9. Kiyozumi D, Noda T, Yamaguchi R, Tobita T, Matsumura T, Shimada K, et al. NELL2-mediated lumicrine signaling through OVCH2 is required for male fertility. Science. 2020;368:1132–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kiyozumi D, Shimada K, Chalick M, Emori C, Kodani M, Oura S, et al. A small secreted protein NICOL regulates lumicrine-mediated sperm maturation and male fertility. Nat Commun. 2023;14:2354.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kiyozumi D. The molecular mechanisms of mammalian sperm maturation regulated by NELL2-ROS1 lumicrine signaling. J Biochem [Internet]. 2022;172:341–6.

    Article  CAS  PubMed  Google Scholar 

  12. Kiyozumi D. New insights into lumicrine secreted signalling that regulates the function of the mammalian male reproductive tract. Dev Biol. 2023;503:111–2.

    Article  CAS  PubMed  Google Scholar 

  13. Kiyozumi D. Lumicrine signaling: extracellular regulation of sperm maturation in the male reproductive tract lumen. Genes Cells. 2023;28:757–63.

    Article  CAS  PubMed  Google Scholar 

  14. Sonnenberg-Riethmacher E, Walter B, Riethmacher D, Gödecke S, Birchmeier C. The c-ros tyrosine kinase receptor controls regionalization and differentiation of epithelial cells in the epididymis. Genes Dev [Internet]. 1996;10:1184–93.

    Article  CAS  PubMed  Google Scholar 

  15. Jun HJ, Roy J, Smith TB, Wood LB, Lane K, Woolfenden S, et al. ROS1 signaling regulates epithelial differentiation in the epididymis. Endocrinology. 2014;155:3661–73.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Sullivan R, Mieusset R. The human epididymis: its function in sperm maturation. Hum Reprod Update. 2016;22:574–87.

    Article  CAS  PubMed  Google Scholar 

  17. Sullivan R, Légaré C, Lamontagne-Proulx J, Breton S, Soulet D. Revisiting structure/functions of the human epididymis. Andrology. 2019;7:748–57.

    Article  CAS  PubMed  Google Scholar 

  18. Robertson MJ, Kent K, Tharp N, Nozawa K, Dean L, Mathew M, et al. Large-scale discovery of male reproductive tract-specific genes through analysis of RNA-seq datasets. BMC Biol. 2020;18:103.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hermann BP, Cheng K, Singh A, Roa-De La Cruz L, Mutoji KN, Chen I-C, et al. The mammalian spermatogenesis single-cell transcriptome, from Spermatogonial Stem cells to Spermatids. Cell Rep. 2018;25:1650–1667e8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kiyozumi D, Ikawa M. ADGRG2 is dispensable for lumicrine signalling regulating epididymal initial segment differentiation and gene expression. Biol Reprod. 2023;109:474–81.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Schiweck W, Buxbaum B, Schätzlein C, Neiss HG, Skerra A. Sequence analysis and bacterial production of the anti-c-myc antibody 9E10: the V(H) domain has an extended CDR-H3 and exhibits unusual solubility. FEBS Lett. 1997;414:33–8.

    Article  CAS  PubMed  Google Scholar 

  22. Robaire B, Hinton BT, Orgebin-Crist M-C. The epididymis. Knobil and Neill’s Physiology of Reproduction. 2006;1071–148.

  23. Kobayashi K, Endo T, Matsumura T, Lu Y, Yu Z, Matzuk MM, et al. Prss55 but not Prss51 is required for male fertility in mice†. Biol Reprod. 2020;103:223–34.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Sakurai N, Fujihara Y, Kobayashi K, Ikawa M. CRISPR/Cas9-mediated disruption of lipocalins, Ly6g5b, and Ly6g5c causes male subfertility in mice. Andrology. 2022. https://doi.org/10.1111/andr.13350.

    Article  PubMed  Google Scholar 

  25. Da Ros VG, Maldera JA, Willis WD, Cohen DJ, Goulding EH, Gelman DM, et al. Impaired sperm fertilizing ability in mice lacking Cysteine-RIch secretory protein 1 (CRISP1). Dev Biol. 2008;320:12–8.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Marcello MR, Jia W, Leary Ja, Moore KL, Evans JP. Lack of tyrosylprotein sulfotransferase-2 activity results in altered sperm-egg interactions and loss of ADAM3 and ADAM6 in epididymal sperm. J Biol Chem. 2011;286:13060–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhou YS, Webb S, Lettice L, Tardif S, Kilanowski F, Tyrrell C, et al. Partial deletion of chromosome 8 β-defensin cluster confers sperm dysfunction and infertility in male mice. PLoS Genet. 2013;9:e1003826.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Zhang C, Zhou Y, Xie S, Yin Q, Tang C, Ni Z, et al. CRISPR/Cas9-mediated genome editing reveals the synergistic effects of β-defensin family members on sperm maturation in rat epididymis. FASEB J. 2018;32:1354–63.

    Article  CAS  PubMed  Google Scholar 

  29. Cheng CY, Mruk DD. The blood-testis barrier and its implications for male contraception. Pharmacol Rev. 2012;64:16–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Légaré C, Sullivan R. Expression and localization of c-ros oncogene along the human excurrent duct. Mol Hum Reprod. 2004;10:697–703.

    Article  PubMed  Google Scholar 

Download references

Funding

This work was supported in part by the Ministry of Education, Culture, Sports, Science and Technology (MEXT)/Japan Society for the Promotion of Science (JSPS) KAKENHI grants (JP21H02487, JP21H00231, and JP21K19263), Japan Science and Technology Agency (JPMJPR2143), the Japan Foundation for Applied Enzymology (2023-10), the Chugai Foundation for Innovative Drug Discovery Science (2022-I-05), and the UBE Foundation to DK.

Author information

Authors and Affiliations

Authors

Contributions

D.K. designed and performed experiments. D.K. wrote and revised the manuscript.

Corresponding author

Correspondence to Daiji Kiyozumi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Declaration of generative AI in scientific writing

The author declares that no generative AI was used in preparing the manuscript.

Submission declaration and verification

The author declares that this manuscript has not been published previously and is not under consideration for publication elsewhere.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kiyozumi, D. Expression of NELL2/NICOL-ROS1 lumicrine signaling-related molecules in the human male reproductive tract. Reprod Biol Endocrinol 22, 3 (2024). https://doi.org/10.1186/s12958-023-01175-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12958-023-01175-6

Keywords