Next Article in Journal
Continuous Glucose Monitoring (CGM) and Metabolic Control in a Cohort of Patients with Type 1 Diabetes and Coeliac Disease
Next Article in Special Issue
Anti-Dyslipidemic and Anti-Diabetic Properties of Corosolic Acid: A Narrative Review
Previous Article in Journal
Unveiling Thyroid Disease Associations: An Exceptionality-Based Data Mining Technique
Previous Article in Special Issue
Role of the Androgen Receptor in Gender-Related Cancers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hormones and Signaling Pathways Involved in the Stimulation of Leydig Cell Steroidogenesis

by
Karine de Mattos
1,†,
Kenley Joule Pierre
1,† and
Jacques J. Tremblay
1,2,*
1
Reproduction, santé de la mère et de l’enfant, Local T3-67, Centre de Recherche du CHU de Québec–Université Laval, 2705 boul. Laurier, Quebec, QC G1V 4G2, Canada
2
Centre for Research in Reproduction, Development and Intergenerational Health, Department of Obstetrics, Gynecology, and Reproduction, Faculty of Medicine, Université Laval, Quebec, QC G1V 0A6, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Endocrines 2023, 4(3), 573-594; https://doi.org/10.3390/endocrines4030041
Submission received: 1 July 2023 / Revised: 22 July 2023 / Accepted: 28 July 2023 / Published: 1 August 2023
(This article belongs to the Special Issue Feature Papers in Endocrines 2023)

Abstract

:
Leydig cells, located in the testis interstitial space, are the primary source of testosterone in males. Testosterone plays critical roles in both reproductive and metabolic functions and therefore is essential for male health. Steroidogenesis must be properly regulated since dysregulated hormone production can lead to infertility and metabolic disorders. Leydig cell steroidogenesis relies on the coordinated interaction of various factors, such as hormones and signaling molecules. While luteinizing hormone (LH) is the main regulator of Leydig cell steroidogenesis, other molecules, including growth hormones (GH), prolactin, growth factors (insulin, IGF, FGF, EGF), and osteocalcin, have also been implicated in the stimulation of steroidogenesis. This review provides a comprehensive summary of the mechanisms and signaling pathways employed by LH and other molecules in the stimulation of Leydig cell steroidogenesis, providing valuable insights into the complex regulation of male reproductive and metabolic health.

1. Introduction

Steroid hormones play a vital role in various physiological processes, including growth and development, reproduction, and metabolism. Steroid hormones are synthesized by different endocrine glands and organs, and their secretion is tightly regulated to maintain homeostasis. Among the organs involved in steroid hormone synthesis, the testes play a crucial role in male physiology through the production of androgens by Leydig cells. Leydig cells, located in the testis interstitial compartment, are responsible for the production and secretion of testosterone and insulin like-3 (INSL3), two critical hormones for reproductive function and overall health in men. In addition to regulating male fertility, testosterone also plays essential roles in the development of secondary sexual characteristics, muscle mass, bone density, and red blood cell production (reviewed in [1]).
The homeostasis of steroidogenesis is of paramount importance for male health. Testosterone deficiency or excess can result in differences of sex development (DSD), as well as other pathologies in men, including infertility, erectile dysfunction, osteoporosis, and metabolic disorders. Testosterone synthesis and secretion in males is therefore tightly regulated by multiple signaling pathways, and not surprisingly, understanding these pathways remain a topic of active research.
In contrast to endocrine cells that produce polypeptide hormones, steroidogenic cells store minimal amounts of steroid hormones and lack secretory vesicles for rapid release (reviewed in [2]). Therefore, a prompt steroidogenic response involving the coordinated action of several pathways is required for rapid de novo synthesis of steroid hormones.
This review provides a comprehensive summary of the current state of knowledge of signaling pathways involved in the stimulation of Leydig cell steroidogenesis.

2. Main Factors Involved in the Stimulation of Leydig Cells

2.1. Luteinizing Hormone

Luteinizing hormone (LH) belongs to the glycoprotein hormone family, which also comprises follicle-stimulating hormone (FSH) and thyroid-stimulating hormone (TSH) [3]. LH is a glycoprotein heterodimer composed of an alpha and beta subunit. While the alpha subunits of LH, FSH, and TSH are identical, the beta subunit of each hormone is distinct and confers its unique biological effects [3].
Gonadotrope cells located in the anterior pituitary gland synthesize and secrete LH in a pulsatile manner due to hypothalamic GnRH, which is also secreted in a pulsatile manner. LH, along with FSH and placental chorionic gonadotropin (CG), are classified as gonadotropins due to their effects on the gonads. LH is a crucial component of the hypothalamic-pituitary-gonadal (HPG) axis that links the nervous system with the gonads. The HPG axis is controlled by a classic negative feedback loop where circulating testosterone secreted by the testes continuously feeds back to the hypothalamus and pituitary to adjust GnRH and LH output. In males, the LH receptor is primarily present in Leydig cells. Although CG is exclusively found in primates and equids, the LH receptor recognizes both LH and CG and is thus referred to as the luteinizing hormone/chorionic gonadotropin receptor (LHCGR) (reviewed in [4]). The LHCGR receptor, along with other glycoprotein hormone receptors, belongs to the G protein-coupled receptor superfamily. In Leydig cells, the binding of LH to LHCGR induces a conformational change in the receptor, initiating a signaling cascade that ultimately results in increased testosterone synthesis.
Multiple mouse models have been utilized to elucidate the functions of LH in Leydig cells, including GnRH-deficient mice and knockout mice for LH and LHCGR. GnRH-deficient hypogonadal (hpg) mice have significantly decreased pituitary and plasma gonadotropins, which directly affect the reproductive system. This condition results in the development of cryptorchidism, accompanied by underdeveloped testes and a significantly diminished number of Leydig cells (approximately 10% of normal values), and ultimately infertility [5,6]. Similarly, Lhb and Lhcgr knockout mice are infertile, have reduced levels of serum and intratesticular testosterone, smaller testes and accessory glands, and possess only a few Leydig cells [7,8,9,10] (reviewed in [11]). In addition, Lhcgr knockout mice exhibit cryptorchidism and increased serum LH levels [7,8]. Sexual differentiation and fetal gonadal development are, however, normal in the knockout models, indicating that, unlike humans, fetal testosterone production required for masculinization is not dependent on gonadotropins in mice.
Naturally occurring mutations and polymorphisms in the LHB and LHCGR genes have been identified in humans (reviewed in [12]). Most mutations are in LHCGR and only a few in LHB (reviewed in [11,12]). Inactivating mutations in LHCGR lead to a failure of Leydig cell differentiation, resulting in Leydig cell hypoplasia (LCH). Males with inactivating mutations in LHB are normally masculinized at birth but later present delayed or lack of spontaneous puberty that is accompanied by hypogonadism, low testosterone levels, and infertility (reviewed in [11,12]). As expected, the phenotype of the homozygous Lhb knockout male mice closely mimics that of humans harboring inactivating LHB mutations [9] (reviewed in [12]). These findings indicate that LH participates actively in the control of Leydig cell differentiation, steroidogenesis, and male fertility.
The binding of LH to LHCGR on the surface of Leydig cells activates multiple pathways, which trigger downstream signaling cascades via G-proteins (Figure 1). In the text that follows, we provide an overview of the primary pathways downstream of LH that promote Leydig cell steroidogenesis, which include PKA, CAMKI, ERK1/2, PKC and PKB/AKT.

2.1.1. cAMP-Dependent Protein Kinase

Activation of LHCGR/G-proteins stimulates adenylate cyclase (AC), which increases the conversion of ATP to cyclic AMP (cAMP). cAMP acts as a second messenger, activating cAMP-dependent protein kinase A (PKA). This signaling pathway is commonly known as the Gs/AC/cAMP/PKA pathway (reviewed in [13]). PKA is a ubiquitous serine/threonine protein kinase that recognizes a consensus sequence RRXS*/T*Hpo in target proteins, where “*” represents the phosphorylation site and “Hpo”, a hydrophobic residue [14] (reviewed in [15]).
Since its discovery in 1968 [16], PKA has been linked to several cellular functions such as growth and cell division [17], metabolism and regulation of energy balance (reviewed in [18]), cell differentiation [19], transcriptional regulation [20], and spermatogenesis and sperm motility [21]. In an inactive state, PKA exists as a tetrameric holoenzyme composed of two regulatory subunits (R) bound to two catalytic subunits (C) (reviewed in [15]). The binding of cAMP to the PKA regulatory subunits leads to the release of the catalytic subunits from the regulatory subunits [22,23]. Over the years, two different forms of PKA, referred to as PKA type I and type II, have been identified. These isoforms share the same catalytic subunit but possess different regulatory subunits (RI and RII) [24].
In mice, inactivation of the Prkaca gene, which encodes the catalytic alpha subunit (Cα) of PKA, leads to partial lethality, with less than 30% of animals surviving to adulthood [25]. While testis size in these mice appears normal compared to their body weight, they nonetheless exhibit a reduction in sperm count and an increase in abnormal spermatozoa [25]. In humans, around 65% of Carney complex (CNC) tumor patients exhibit haploinsufficiency of the RIα gene (PRKAR1A), which correlates with a notable decrease in fertility among male CNC patients [26,27].
In the mouse MA-10 Leydig cell line, active PKA translocates to the nucleus, where it phosphorylates multiple transcription factors, such as GATA4 and bZIP family members. Phosphorylation of GATA4 at Ser261 leads to increased GATA4-dependent activation of several gene promoters such as Star, Cyp17a1, aromatase, and Inha (inhibin α) [28,29]. More recently, we have shown that PKA also cooperates with COUP-TFII (NR2F2) and STAT5B to activate the Star promoter in MA-10 Leydig cells [29].

2.1.2. Calcium

LHCGR activation in Leydig cells triggers the production of inositol 1,4,5-triphosphate (IP3), which in turn binds to its receptor (IP3R), resulting in the release of calcium (Ca2+) [30]. Ca2+ release alters cytosolic Ca2+ concentration, playing a crucial role in facilitating steroidogenesis. Calcium serves as a key second messenger, controlling various cellular functions such as autophagy, apoptosis, and cell proliferation (reviewed in [31]). The levels of cytosolic Ca2+, directly and indirectly, regulate the activity of proteins and enzymes. Cells actively store Ca2+ at high concentrations, particularly in the endoplasmic reticulum (ER), due to its essential role in intracellular processes (reviewed in [32]). The release of Ca2+ from the ER is facilitated by the ryanodine receptor (RyR) and the IP3R, acting as the primary channels responsible for this process [30]. These channels rapidly release Ca2+ into the cytoplasm, providing the cell with the necessary Ca2+ for the execution of various intracellular processes, including fertilization, gene transcription, muscle contraction, exocytosis and cell differentiation, proliferation and motility (reviewed in [33]). Extracellular Ca2+ can also enter through plasma membrane channels, thus serving as an additional source of Ca2+ for the cell.
Studies using rat Leydig cells have revealed that both LH and dibutyryl cyclic AMP (db-cAMP), an analog of cAMP, increase intracellular Ca2+ concentration through extracellular and intracellular sources [34,35]. Additionally, stimulation of MA-10 Leydig cells with forskolin (Fsk) increases intracellular Ca2+ levels, resulting in enhanced steroid production [36]. Conversely, inhibition of RyRs in these cells leads to reduced steroidogenesis. This inhibition specifically impacts the activity of the cAMP-induced Star promoter, leading to a decrease in STAR production and subsequently affecting the translocation of cholesterol from the outer to the inner mitochondrial membrane [36].
Calmodulin (CaM), a protein highly responsive to Ca2+ levels, serves as an upstream activator for several calcium-dependent proteins, including calcium/calmodulin-dependent protein kinases I, II and IV (CAMKI, CAMKII, and CAMKIV) (reviewed in [37]). Among the calcium/calmodulin-dependent protein kinases, CAMKI has been identified as the main CAMK that significantly influences steroidogenesis in MA-10 Leydig cells [38].

Calcium/Calmodulin-Dependent Protein Kinase I

CAMKI, the smallest member of the Ca2+/CaM-dependent protein kinase family, is a versatile serine/threonine-specific multifunctional protein kinase. It recognizes a consensus sequence BXRXX(S/T)XXXB, where B represents a hydrophobic amino acid, and X can be any amino acid [39]. Structurally, CAMKI is a monomeric enzyme consisting of an N-terminal catalytic domain and a C-terminal regulatory domain. The N-terminal domain includes the ATP-binding domain (residue 1–100) and the substrate-binding domain (residue 101–275). In the absence of Ca2+, CAMKI remains in an autoinhibited state due to the folding of the regulatory domain over the catalytic domain (reviewed in [40]). The key factor governing the maximal activation of CAMKI involves two crucial steps. First, there must be an elevation in the intracellular concentration of Ca2+. Second, CAMKI kinase (CAMKKI) phosphorylates a specific residue, Thr177, in CAMKI [41]. These two events act as limiting factors in achieving peak CAMKI activation.
CAMKI is the most recently identified kinase in Leydig cells and has emerged as a subject of interest in recent studies, particularly in MA-10 Leydig cells. Although our understanding of CAMKI action in Leydig cell function is still in its early stages, emerging evidence suggests it is involved in crucial cellular processes. Previous studies have revealed that CAMKI acts as an activator, collaborating with multiple proteins, including transcription factors, to enhance the expression of genes associated with steroid hormone production [29] (reviewed in [42,43]). Some of the transcription factors found to cooperate with CAMKI include members of the nuclear receptor family NR4A (NUR77/NR4A1, NURR1/NR4A1, NOR1/NR4A3), NRF2 members (COUP-TFI/NR2F1 and COUP-TFII/NR2F2), GATA4, STAT5B, CREB, and cJUN. These findings highlight the multifunctional nature of CAMKI and its potential role in regulating steroidogenesis downstream of LHCGR activation in Leydig cells.

2.1.3. Protein Kinase C

Protein kinase C (PKC) is another important kinase that is activated by the binding of LH/hCG to its receptor. One of the signaling pathways triggered by LHCGR activation involves phospholipase C (PLC). Through the process of hydrolysis, PLC breaks down phosphatidylinositol 4,5-bisphosphate (PIP2), resulting in the release of diacylglycerol (DAG) and IP3 [44]. DAG plays a vital role as a potent activator of PKC, while IP3 leads to an increase in Ca2+ levels, as mentioned above (see calcium section).
PKC is a family of serine/threonine protein kinases that are involved in several cellular functions, including cell proliferation and differentiation (reviewed in [45]), autophagy and apoptosis [46], and regulation of chloride channel in the human kidney [47].
The PKC family comprises several isozymes that are classified into four subfamilies based on their activation mechanism: cPKC, nPKC, aPKC and PKN [48]. Each subfamily shares a common structural organization consisting of a regulatory domain, a highly conserved catalytic domain, and variable regions (reviewed in [48,49,50]). Importantly, all PKC isozymes recognize the consensus phosphorylation sites (R/K)X(S/T), (R/K)(R/K)X(S/T), (R/K)XX(S/T), (R/K)X(S/T)(R/K), and (R/K)XX(S/T)XR/K in target proteins (reviewed in [50]).
The conventional or classic cPKC subfamily proteins consist of cPKCα, βI, βII, and γ, which are activated by DAG, phosphatidylserine, and Ca2+. In contrast, the novel or non-classic nPKC subfamily proteins include nPKCδ, ε, η, and θ, which are activated by DAG and phosphatidylserine but not Ca2+ due to the absence of Ca2+ coordinating residues. The atypical subfamily of aPKC proteins comprises aPKCζ, ι, and λ. These aPKC members contain a PBI domain that confers specificity in intracellular signaling by interacting with scaffold proteins. Unlike other subfamilies, aPKC kinases are not activated by DAG, as their C1 domain does not bind DAG. Lastly, the PKN subfamily includes PKN1, PKN2, and PKN3, which all contain an HRI domain that mediates their activation by Rho proteins (reviewed in [48,49,50]).
MA-10 Leydig cells express several PKC isozymes, including PKCα, βI, βII, δ, γ, ε, θ and ζ [51,52]. Furthermore, studies in MA-10 Leydig cells have shown that phorbol 12-myristate 13-acetate (PMA), an analog of DAG, can increase the levels and phosphorylation of PKCα, δ, and ε, which was also associated with an increase in the level of STAR protein and progesterone production [51,53]. In R2C Leydig cells, inhibition of PLC and PKC leads to a significant decrease in steroid production and phosphorylation of cAMP-responsive element binding (CREB) [54]. This decrease is accompanied by a reduction in Star gene transcription [54]. The PKC pathway also influences the orphan nuclear receptor DAX1/NR0B1, which modulates Star expression [55].

2.1.4. Extracellular Signal-Regulated Kinase

Extracellular signal-regulated kinase 1 and 2 (ERK1 and ERK2) are serine/threonine kinases, members of the MAP kinase (MAPK) signaling cascade. This pathway is involved in many cellular functions such as growth, proliferation, cell differentiation, mitosis, metabolism and apoptosis (reviewed in [56,57]). Although ERK1 and ERK2 have minor differences, they share numerous functions and are collectively referred to as ERK1/2. The activation of ERK1/2 is mediated through a sequential cascade of reactions known as the Ras/Raf/MEK/ERK1/2 signaling pathway. In response to extracellular stimuli, Ras is activated, subsequently activating the MAPKKK Raf. This, in turn, triggers the activation of two MAPKKs, MEK1 and MEK2, which phosphorylate the MAPKs ERK1/2. Once activated, ERK1/2 translocates to the nucleus, where it phosphorylates and activates transcription factors that influence gene expression, recognizing preferentially a consensus sequence PXS/TP (reviewed in [58,59]).
Several studies have explored the role of ERK1/2 in Leydig cells, revealing its involvement in steroidogenesis and the proliferation of postnatal Leydig cells [55,60,61] (reviewed in [62]).
A Cyp17a1-iCre mouse line, which expresses the Cre recombinase in steroidogenic tissues, including Leydig cells, was utilized to conditionally inactivate Mek1 and Mek2 [61,63]. MEK1/MEK2-deficient male mice exhibit a reduction in the number of Leydig cells compared to control animals. Moreover, testosterone levels upon hCG stimulation are reduced in these mice, along with a significant decrease in the expression of key steroidogenic genes, such as Star, Hsd3b6, Cyp17a1 and Hsd17b3 [61,63]. These findings demonstrate that ERK1/2 plays a pivotal role in the regulation of steroidogenesis.
In various Leydig cell models, including MA-10, MLTC-1, and primary rat Leydig cells, it has been observed that activation of LHCGR induces Ras activation, subsequently leading to the phosphorylation of ERK1/2 [64,65]. In addition, treatment of MA-10 and MLTC-1 Leydig cells with two different MEK inhibitors, U0126 and PD98059, significantly reduces LH-induced steroidogenesis while increasing STAR protein levels, but not its phosphorylation [55,65].
In MA-10 Leydig cells, ERK1/2 has been found to functionally cooperate with STAT5B and GATA4 to activate the Star promoter [29]. The ERK1/2-GATA4 cooperation on Star is consistent with a study conducted in rat primary cardiomyocyte cells, which revealed that ERK1/2 phosphorylates GATA4 at Ser105 [66]. Interestingly, mice carrying a GATA4 S105A mutation exhibit a substantial decrease in plasma and intratesticular testosterone levels, indicating the crucial role of this phosphorylation in the regulation of testosterone [67].

2.1.5. Protein Kinase B

Following LHCGR activation, another kinase called protein kinase B (PKB), also known as AKT is activated. PKB is a widely expressed serine/threonine protein kinase that comprises three isoforms: PKBα/AKT1, PKBβ/AKT2, and PKBγ/AKT3 [68,69,70,71]. The PKB/AKT isoforms phosphorylate target proteins at a consensus sequence known as RXRXX(S/T) (reviewed in [72]). Since its discovery, PKB/AKT has been associated with several cellular processes, including glucose metabolism, cell proliferation, apoptosis, gene transcription, and cell migration (reviewed in [72]). The activation of PKB/AKT depends on the upstream kinase phosphatidylinositol-3-kinase (PI3K), a lipid kinase that phosphorylates PKB/AKT on Thr308 and Ser473 [73].
In MA-10 Leydig cells, hCG stimulation leads to the activation of PKB/AKT, which is associated with increased levels of several genes and proteins important for steroidogenesis, including STAR, JUNB, and NUR77/NR4A1 [74]. Consistent with this, activation of PKB/AKT in MA-10 Leydig cells results in increased production of steroid hormone [74].

2.2. Growth Hormone (GH)

In addition to LH, other hormones are known to stimulate Leydig cell steroidogenesis (Figure 2), including growth hormone (GH). GH is a crucial peptide hormone that regulates several essential physiological processes in the body. It is primarily secreted by somatotrope cells located in the anterior pituitary, but local production of GH by many tissues has also been reported (reviewed in [75]). GH mediates its effects by binding to the GH-receptor (GHR) and, in some species, including humans, to the prolactin receptor (PRLR) [76,77] (reviewed in [78]).
In contrast to dimeric glycoproteins such as gonadotropins, two receptors are needed to establish a trimeric structure composed of two membrane receptors and the GH molecule (reviewed in [79]). While the majority of human GHR is found in the liver, it is also abundant in all cellular components of the human reproductive system [80]. In males, GHR and GH binding proteins are present in the testes (Leydig and Sertoli cells), seminal vesicles, epididymis, vas deferens, and prostate [80].
GH acts both directly and indirectly to induce anabolic and metabolic responses in multiple target tissues. Directly, GH acts via the GHR, while indirectly, GH stimulates the production of insulin-like growth factor 1 (IGF1) not only in the liver but also in peripheral target tissues (reviewed in [78,81]).
Studies using animal models have revealed the importance of GH in male reproductive health (reviewed in [81]). GH-deficient male rats and mice exhibit smaller testes, underdeveloped secondary sex organs, delayed puberty, and reduced fertility rates [82,83,84,85]. In addition, male mice lacking GH have lower intratesticular testosterone levels and their ability to produce testosterone in response to LH is also diminished [82,83,84]. Stimulation of Leydig cells with GH increases Star gene expression and testosterone production [86,87,88]. Moreover, GH improves Leydig cell responsiveness to physiological hCG concentration (reviewed in [89]). In prepubertal male rats, treatment with recombinant hGH results in an increase in body weight, early onset of puberty, activation of spermatogenesis, Leydig cell differentiation and testosterone production [90]. These findings suggest that in the testis, GH mediates its effects, at least in part, by acting directly on Leydig cells.

Janus Kinase

Janus kinase (JAK) is a distinct family of tyrosine kinases that comprises four members: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2). In mammals, JAK1, JAK2, and TYK2 are ubiquitous, while JAK3 is found mainly in bone marrow, the lymphatic system, endothelial cells, and vascular smooth muscle cells (reviewed in [91]). Members of the JAK family exhibit a unique structure with more than one kinase domain, contributing significantly to their functional versatility and regulatory capabilities (reviewed in [92]). In mice, inactivation of Jak1 or Jak2 leads to embryonic lethality, emphasizing their critical roles [93,94,95].
In the canonical pathway, binding of GH to GHR leads to receptor activation and formation of GHR dimers. This triggers the transphosphorylation of JAKs, which subsequently phosphorylate tyrosine residues on the bound receptor, creating a docking site for members of the Signal Transducers and Activators of Transcription (STATs) family of transcription factors. Upon docking, JAKs phosphorylate STATs, which then dissociate from the receptor. Phosphorylated STATs dimerize and translocate to the nucleus to regulate gene transcription. STATs bind to the promoter region of target genes, specifically to the γ-interferon-activated sequence (GAS; TTCNNNGAA) (reviewed in [91,96]).
Among the STAT family members, STAT5A and STAT5B have been identified in Leydig cells [97]. In MA-10 Leydig cells, GH treatment induces phosphorylation of STAT5B, facilitating its translocation to the nucleus [97]. A recent study conducted in MA-10 Leydig cells showed that activated-STAT5B increases Star transcription by directly binding to a GAS sequence and by cooperating with cJUN [87]. In addition, activated-STAT5B activates the Nr4a1/Nur77 promoter in MA-10 Leydig cells [87]. Consistent with this, inhibition of JAK by tofacitinib in Leydig cells decreases STAT5B phosphorylation [98].

2.3. Prolactin

Prolactin (PRL) is a peptide hormone that is predominantly produced and secreted by lactotrope cells of the anterior pituitary gland. The structural similarities between the PRL and GH genes and receptors suggest a shared evolutionary origin, likely derived from a common ancestral precursor [99]. The role of PRL in females has been extensively studied and is mainly associated with lactation and mammary gland development. In the context of male reproductive physiology, several studies have provided evidence for the involvement of PRL, particularly in Leydig cell steroidogenesis (reviewed in [100]). However, it is important to acknowledge that there is conflicting data regarding the precise role of PRL in Leydig cells, which can be attributed to the functional state of Leydig cells and the timing and dosage of PRL exposure.
In vitro studies have shown that PRL plays a significant role in male reproduction by potentially stimulating testicular steroidogenesis. PRL indirectly contributes to testosterone production in Leydig cells by modulating the release of gonadotropins from the pituitary gland (reviewed in [100]). PRL is also thought to directly regulate steroidogenesis by increasing the number of LH receptors in Leydig cells and enhancing the sensitivity of these cells to LH stimulation [101,102,103,104,105]. For instance, when plasma PRL levels are reduced, LH receptor levels in rat Leydig cells are also reduced [106,107,108]. In addition, in MA-10 Leydig cells, the influence of PRL on hCG-induced steroidogenesis was found to be biphasic; at low concentrations of PRL, steroidogenesis is stimulated, whereas, at high concentrations of PRL, steroidogenesis is inhibited [109]. On the other hand, genetic deletion of either the hormone (PRL-KO) or its receptor (PRLR-KO) in the mouse had no impact on male reproductive functions [110,111]. This lack of male reproductive phenotype could be due to compensation by other hormones, cytokines, or homologous receptors (reviewed in [100]).
The membrane receptor for PRL, PRLR, is present in Leydig cells across various species, including humans, mice, rats, and rams [112,113,114,115]. However, in humans, it appears that Leydig cells do not exhibit detectable PRL binding to interstitial cells [116]. Similar to GHR, activation of PRLR involves ligand-induced sequential receptor dimerization (reviewed in [117]). Upon PRL binding to PRLR and subsequent dimerization, a signaling cascade is initiated with the activation of JAK2, which in turn phosphorylates STAT5A and STAT5B (Figure 2). As described above, for GH action, phosphorylated STAT5B then translocates to the nucleus, where it activates gene transcription (reviewed in [117]). STAT5 was found to be involved in PRL signaling in the MA-10 Leydig cell line. However, the role of STAT5 in steroidogenesis in primary Leydig cell cultures depends on the developmental status of Leydig cells. While STAT5 is not involved in PRL signaling in primary Leydig cell cultures from juvenile rats [97], in primary Leydig cell cultures from adult rats, PRL increases STAT5 phosphorylation and Lhcgr mRNA levels [98]. Conversely, the JAK inhibitor tofacitinib reduces PRL-mediated phosphorylated STAT5 and Lhcgr mRNA levels in primary Leydig cell cultures from adult rats [98]. PRL also regulates Ca2+ uptake in Leydig cells via increased Ca2+ entry [118]. It is well known that free calcium activates steroidogenesis via the CAMKI pathway, as described above. Therefore, it is likely that PRL regulates steroidogenesis via both the JAK/STAT and the CAMKI pathways.

2.4. Growth Factors

Several growth factors also contribute to Leydig cell steroidogenesis, including insulin, insulin-like growth factor (IGF), fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), transforming growth factor (TGF)-β, TGFα, as well as IGF-binding proteins (IGFBPs). In the following sections, the roles and contributions of a subset of these factors that have been widely studied in Leydig cells is summarized (Figure 2).

2.4.1. Insulin Family of Growth Factors

The insulin family of growth factors, including insulin, insulin-like growth factor 1 (IGF1), IGF2, and relaxin, are small polypeptides that play critical roles in controlling growth, metabolism, and reproductive functions. Among them, insulin and IGF1 have been extensively studied in the context of testicular function. Insulin and IGF1 actions are mediated through the activation of two related tyrosine kinase receptors, insulin receptor (INSR) and IGF1 receptor (IGF1R). These receptors are composed of two extracellular α subunits and two transmembrane β subunits, which come together to form heterotetrameric glycoproteins (reviewed in [119]). Interestingly, both insulin and IGF1 can bind to both receptors, albeit with different affinities. Insulin exhibits a high affinity for INSR but can also bind to IGF1R, albeit with a lower affinity. Similarly, IGF1 exhibits preferential binding to IGF1R while also displaying a lower affinity for INSR (reviewed in [119]).

Insulin

Insulin is a hormone produced primarily by the β-cells of the pancreas in response to elevated blood glucose levels [120] (reviewed in [121]). It acts by binding to its receptor INSR present in several tissues, leading to lower blood glucose levels by promoting cellular uptake and utilization of glucose. Insulin also stimulates the synthesis and storage of glycogen and fat and inhibits the breakdown of stored glycogen, fat, and protein in muscles (reviewed in [121]). Insulin deficiency affects the entire metabolism, including male fertility [122]. The role of insulin in the regulation of metabolic processes is often investigated in patients with type 1 diabetes, who produce very little or no insulin, and type 2 diabetes, characterized by insulin resistance.
Several animal models were developed to investigate the molecular mechanisms and histopathological processes involved in insulin function, including spontaneously diabetic rodent strains, streptozotocin (STZ)-induced diabetes, Ins2 and Insr knockout mice, among others [123,124,125,126]. In diabetic rats, Leydig cell function is impaired, resulting in lower testosterone levels [124,126,127], a condition partially reversed by insulin treatment [126]. Similarly, male patients with type 2 diabetes exhibit significantly lower serum INSL3 concentrations [128]. This is consistent with data from diabetic rats where lower INSL3 levels are also observed, levels that are increased in insulin-treated rats [126]. In mice, double knockout of Insr and Igf1r results in a dramatic reduction in the number of Leydig cells that are unresponsive to hCG stimulation, while steroidogenesis in individual knockout male mice is not affected [129]. This indicates a functional redundancy between the two receptors and pathways in post-natal Leydig cells.
INSR is present in Leydig cells, and the addition of insulin to Leydig cells in primary culture and to TM3 Leydig cells increases steroidogenesis [130,131]. Conversely, several studies have reported the negative impact of insulin deficiency on Leydig cells [124,125,126]. Very high levels of insulin, such as those observed in obesity and type 2 diabetes, have negative effects on steroidogenesis through the induction of the atypical nuclear receptor DAX1/NR0B1, which represses hormone-induced steroidogenesis [132].
Insulin triggers a series of events upon binding to its tyrosine kinase receptor INSR, resulting in the activation of at least two pathways, PI3K/PKB and Raf/Ras/MEK/ERK1/2 (reviewed in [133]). In Leydig cells, both pathways appear to play a role in mediating insulin action. However, the exact mechanism of insulin action in the regulation of Leydig cell steroidogenesis remains to be fully characterized.

Insulin-like Growth Factor 1

Insulin-like Growth Factor 1 (IGF1) is a small peptide hormone produced primarily by the liver in response to GH stimulation, but almost all tissues, including the testes, can synthesize IGF1 ([134,135], reviewed in [78]). In the testes, IGF1 and its receptor (IGFR) are present in Sertoli, Leydig, germ, and peritubular cells [119,130,135,136]. In rodent Leydig cells, IGF1 secretion and upregulation of IGFR can be stimulated by LH, hCG, and GH [137,138,139,140].
Several studies involving animal models have shown that IGF1 is essential for male reproduction [141] (reviewed in [78,119,142]). In fact, IGF1-deficient male mice are infertile [141]. Consistent with this, studies have shown that IGF1 is critical for the proliferation, development and functionality of Leydig cells [130,136,143,144]. This growth factor exerts its effects through both para- and autocrine action [143]. Specifically, IGF1 stimulates the proliferation of Leydig cell precursors [136,145] (reviewed in [78]). Conversely, IGF1 deficiency results in altered proliferation and differentiation of Leydig cell precursors, leading to fewer and smaller Leydig cells and lower serum testosterone levels in adulthood [141,146]. As mentioned previously in the section on insulin, inactivation of both Insr and Igf1r leads to a drastic reduction in Leydig cell number and size and steroidogenic failure [129].
Similar to INSR, activated-IGFR leads to the activation of PI3K/PKB and Raf/Ras/MEK/ERK1/2 pathways. In primary cultures of immature rat Leydig cells, IGF1 stimulates the phosphorylation of AKT and ERK1/2 [147,148]. In addition, IGF1 stimulates fetal Leydig cell proliferation through the MEK/ERK1/2 pathway [149].

2.4.2. Fibroblast Growth Factor 9

Fibroblast growth factor 9 (FGF9) belongs to the FGF family, which comprises at least 22 members found in a wide range of cell types (reviewed in [150]). FGF members interact with the extracellular domain of the FGF receptor (FGFR), leading to the activation of the intracellular tyrosine kinase domain, followed by the activation of signaling cascades. Although there are four known FGFRs, there are at least seven functionally distinct receptors due to alternative splicing events [150]. The different isoforms of FGFRs suggest a distinct function in each system. FGF9 is the main FGF family member present in the mammalian testis and binds to FGFR2IIIc, FGFR3IIIb, FGFR3IIIc, and FGFR4 (reviewed in [151]). Within the testes, FGF9 is present in a specific spatiotemporal pattern in Leydig, Sertoli, and germ cells [152,153,154,155]. The presence of FGF9 and its receptors in Leydig cells suggests that FGF9 functions as an autocrine factor in the regulation of Leydig cell steroidogenesis. Indeed, its effects are believed to be primarily mediated through autocrine and paracrine signaling within the testicular microenvironment rather than systemic effects via circulation [156].
Although Fgf9 knockout mice die at birth, analysis of embryos revealed partial or complete XY gonadal sex reversal [157]. A similar phenotype is observed in humans harboring an FGF9 variant (D195N) associated with 46 XY Difference of Sex Development (DSD) [158].
In mouse primary Leydig cells and MA-10 Leydig cells, treatment with FGF9 increases testosterone production [154,156,159], and this involves phosphorylation and activation of AKT, MAPK and PKA pathways [156,159]. Although the downstream targets of FGF9 action in Leydig cells remain to be identified, the importance of the FGF9 pathway for Leydig cell function is well-established.

2.4.3. Epidermal Growth Factor (EGF) Family

The EGF family consists of 11 members divided into four sub-groups based on their receptor binding specificity. Most EGF family members, including EGF, transforming growth factor α (TGFα), amphiregulin (AREG), epigen (EPGN), heparin-binding EGF-like growth factor (HB-EGF), epiregulin (EREG), and betacellulin (BTC), recognize and bind to the EGF receptor (EGFR) also known as ErbB1. Upon ligand binding, EGFR homodimerizes with another EGFR or heterodimerizes with other EGFR/ErbB family members (reviewed in [160,161]). EGF family members have been implicated in the regulation of cell proliferation, differentiation, survival, and motility in several tissues, including the male gonad (reviewed in [160]).
To better understand the function of EGF, numerous mouse knockout models have been developed (reviewed in [161]). While single and even triple (EGF, AREG, TGFα) knockout mice are viable and fertile, disruption of the Egfr gene leads to pre-implantation or post-natal lethality [162,163]. This indicates that the various EGF family members can compensate, at least in part, for the absence of some family members [162,163,164].
Leydig cells of several species, including humans and rodents, contain both EGF and EGFR [165,166]. The impact of EGF on Leydig cell steroidogenesis is controversial. Current data suggest that its effects are subject to various factors, such as cell maturity and experimental conditions, including treatment duration and the cell line used. Several studies reported a stimulatory role for EGF on Leydig cell steroidogenesis. In MA-10 Leydig cells, a time-dependent effect of EGF was observed [167]. Exposure to EGF initially decreases the activity of adenylate cyclase activated by hCG within the first hour. In the next 7 h, EGF stimulates steroidogenesis via a cAMP-independent pathway. Finally, at 8 h post-EGF treatment, a reduction in LHCGR levels is observed. Other studies also support a stimulatory role for EGF in Leydig cell steroidogenesis [148,166,168,169,170,171]. Furthermore, EGFR signaling is required for early hCG-induced steroidogenesis in mouse MLTC-1 Leydig cells, suggesting a crosstalk between EGFR and LHCGR [168]. In addition, male mice treated with an EGF inhibitor (AG1478) exhibit a substantial reduction in serum testosterone levels [168]. Conversely, an inhibitory role for EGF in testosterone production in Leydig cells has also been reported [172]. EGF was found to reduce steroidogenic gene expression and steroid production in progenitor Leydig cells (PLCs) [173]. Moreover, EGF was found to stimulate the proliferation of stem Leydig cells (SLCs) and PLCs while blocking their differentiation into mature adult Leydig cells [173].
Mechanistically, binding of EGF to its receptor EGFR induces phosphorylation of EGFR and ERK1/2 through the extracellular calcium-sensing receptor (CASR). This observation was made in Rice H500 rat non-metastasizing Leydig tumor cells treated with Ca2+. In these tumor cells, activated-CASR increases the production of parathyroid-hormone-related peptide (PTHrP) via multiple signaling pathways, including PKC, MEK, and JNK [174]. In MLTC-1 Leydig cells, treatment with EGF increases testosterone production and STAR protein levels without any significant change in phospho-STAR levels [166]. More recently, LH-mediated EGFR activation was found to activate casein kinase 1α (CK1α), leading to increased testosterone synthesis [175]. Consistent with this, conditional and partial inactivation of the Csnk1a1 gene (encoding CK1α) in steroidogenic cells in the mouse results in a significant reduction in the expression of several steroidogenic genes leading to lower testosterone levels and decreased male fertility [175].
Although these findings support a role for EGF in Leydig cell steroidogenesis, conflicting data on the effects of EGF on Leydig cell function highlights the complexity of this pathway. Additional work is necessary to fully decipher the role of EGF/EGFR in steroidogenesis.

2.5. Vitamin D

Vitamin D (VitD) is a steroid hormone, and its active form 1,25 dihydroxyvitamin D (1,25(OH)2D) is essential for maintaining calcium and phosphate homeostasis and promoting skeletal health (reviewed in [176,177]). In the classical pathway (genomic), VitD binds to the vitamin D receptor (VDR), which forms a heterodimer with the retinoid X receptor (RXR). This VitD/VDR-RXR heterodimer acts as a transcription factor, recognizing vitamin D response elements (VDREs) present in the promoter regions of various target genes (reviewed in [178,179]). VitD also acts through non-genomic pathways involving phospholipases, second messengers, and kinases (reviewed in [178,179]).
While the liver and kidneys are commonly recognized as the primary organs involved in VitD metabolism, the presence of VitD metabolizing enzymes is not confined exclusively to these organs. In tissues other than the liver and kidneys, VitD was shown to have paracrine/autocrine functions (reviewed in [177,180]).
The male reproductive system, including the testis, contains several VitD metabolizing enzymes as well as VDR (reviewed in [178,181]). In the human testis, the enzyme CYP2R1 is predominantly found in Leydig cells [182], where its expression is hCG-dependent [183]. Another essential enzyme, CYP27B1, is also present in Leydig cells. A transgenic reporter mouse model with a 1.5 kb fragment of the human CYP27B1 promoter revealed significant expression in the testis, including Leydig and Sertoli cells [184,185]. VitD action is dependent on the presence of VDR. This receptor is present in Leydig cells and other cells of the male reproductive system, such as Sertoli cells, germ cells, spermatozoa, and epithelial cells (reviewed in [176]). In the human testis, VDR is present in Leydig cells from both the fetal and adult population [186,187,188]. These studies collectively suggest that Leydig cells locally metabolize VitD, indicating an intracrine/autocrine role in testicular function, particularly in steroidogenesis.
Consistent with this, numerous studies have explored the relationship between VitD levels and steroidogenesis (reviewed in [181,189]). For instance, some studies have reported a correlation between decreased 25-hydroxyvitamin D (25-OHD) levels (the immediate precursor of VitD) and lower testosterone concentrations [190,191,192] (reviewed in [189,193,194]). In addition, CYP2R1, CYP27B1, and VDR levels are lower in infertile men compared to those with normal testicular function (reviewed in [181]). A study using testis samples from young men affected by testicular disorders revealed lower levels of 25-OHD compared to those with normal testicular function [182].
To better elucidate the relationship between VitD and male reproduction status, studies using mice models were performed. In CYP27B1- and VDR-deficient mice, reproductive and endocrine function was impaired [195,196,197]. More specifically, in Vdr knockout mice (VdrKO), reproductive performance and sperm quality declined, resulting in smaller litter size, lower live birth rate, and lower number of successful breeding performances [197,198,199,200]. The absence of VDR also affected testes morphology, with smaller adipocytes and reduced lipid droplet accumulation [197]. These results suggest that VDR is a regulator of lipid metabolism and essential for maximal male fertility. Moreover, expression of Hsd3b1 and Cyp11a1 was significantly decreased in the testes of VdrKO mice [197,201]. Interestingly, male rats receiving a VitD-deficient diet for 3 months presented a decrease in the production of testosterone, reduced testis volume, and decreased number of spermatids and spermatocytes, suggesting an essential role of VitD for male testicular function [202]. In the mouse TM3 Leydig cell line, downregulation of VDR results in a notable reduction in the expression of crucial steroidogenic genes, such as Cyp11a1, Hsd3b1, Star, Nr5a1, and Prkaca [197,201]. Conversely, the upregulation of VDR enhances the expression of the majority of these genes. VDR was found to directly regulate Hsd3b1 expression by binding to a VDRE in the proximal promoter region [201]. The modulation of VDR levels also has a significant impact on genes associated with lipid metabolism [197]. Moreover, the knockdown of VDR in TM3 Leydig cells impairs steroid production, further emphasizing its importance in this process [197,201].
Despite the presence of VDR and its associated enzymes in Leydig cells, our understanding of the role and mechanism of VitD action in steroidogenesis remains limited. In addition, there is currently no evidence supporting VitD supplementation for improving testosterone levels (reviewed in [193]). The current literature on the correlation between sex hormone production and systemic changes in VitD levels presents conflicting findings, especially due to compensatory mechanisms (reviewed in [181]). More work is needed to fully comprehend how VitD influences steroid hormone production in Leydig cells.

2.6. Osteocalcin

Osteocalcin (OCN) or bone γ-carboxyglutamic acid protein is a bone-derived factor produced primarily by osteoblasts (reviewed in [203]). Post-translationally, osteocalcin is γ-carboxylated and considered biologically inactive [204]. A fraction of osteocalcin undergoes decarboxylation and reaches the circulation, acting as a hormone. In target tissues, the undercarboxylated (unOCN) form of osteocalcin mainly binds to the G protein-coupled receptor family C group 6-member A (GPRC6A), while in the central nervous system, it binds to GPR158 as well [205,206].
In vivo studies using null mouse models have demonstrated that osteocalcin is essential for male fertility. In fact, osteocalcin knockout mice exhibit reduced testis weight, oligospermia, and low testosterone levels and produce smaller litter sizes than wild-type animals [207]. A similar phenotype was found in Gprc6a knockout mice [205,208]. The correlation between osteocalcin and male reproduction was also investigated in humans (reviewed in [209]). Using populational-based samples and bone disorders patient-based samples, osteocalcin was found to be positively correlated with testosterone serum concentration; patients with lower OCN had lower testosterone levels and vice-versa [210]. Moreover, Oury and collaborators [205] found in a cohort of patients with primary testicular failure two individuals carrying a missense mutation in the Gprc6a locus. Consistent with this, a recent study strongly suggests a role for the unOCN/GPRC6A axis in the regulation of testosterone production [211].
In the testis, the osteocalcin receptor Gprc6a is highly expressed in Leydig cells [208]. The binding of unOCN was shown to regulate steroidogenesis independently of the HPG axis [205]. The GPRC6A receptor is coupled to adenylate cyclase, and once activated, results in increased cAMP production, activation of signaling pathways, and ultimately the activation of downstream effectors such as CREB, which then upregulate the expression of multiple genes encoding essential steroidogenic proteins and enzymes such as STAR, CYP11A1, CYP17A1, and 3βHSD [207] (Figure 2).
unOCN is also implicated in the regulation of vitamin D metabolism in mouse MA-10 Leydig cells, where it was found to stimulate Cyp2r1 gene expression [212], which codes for the CYP2R1 enzyme involved in the conversion of vitamin D into its active form 1,25(OH)2D (see the section on vitamin D).

3. Conclusions

As described in this review, several pathways are involved in the stimulation of Leydig cell steroidogenesis. Recent advances in technology and molecular biology tools (gene editing to easily generate animal models, more efficient genetic screening of patients, development of high throughput functional assays to study receptors/kinases/transcription factors) have led to significant progress in our understanding of the mechanisms and signaling pathways involved. However, further research is needed to fully comprehend the complex interplay between signaling pathways in Leydig cell steroidogenesis. Leydig cell-specific knockout models are a promising tool that will help better understand the roles of specific receptors, signaling cascade proteins, and kinases in the regulation of steroidogenesis. The development and use of new drugs that target these signaling pathways also hold significant potential for the treatment of disorders related to steroid hormone production. Overall, the field of reproductive endocrinology is constantly evolving, and new tools and technologies are leading to a better understanding of the regulation of steroidogenesis.

Author Contributions

K.d.M., K.J.P.: Drafting and revising the manuscript; and final approval of the manuscript to be submitted for publication. J.J.T.: Writing and revising the manuscript; and final approval of the version to be submitted for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the Canadian Institutes of Health Research (CIHR) (funding reference number MOP-81387 and PJT-148738) to J.J.T., K.M. is the recipient of a studentship from the Fonds de recherche du Québec-Santé. K.J.P. is the recipient of a studentship from the Canadian Institutes of Health Research (funding reference number FBD-189317).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest. The figures were created with BioRender.com (accessed on 30 June 2023).

References

  1. Zirkin, B.R.; Papadopoulos, V. Leydig cells: Formation, function, and regulation. Biol. Reprod. 2018, 99, 101–111. [Google Scholar] [CrossRef] [Green Version]
  2. Shen, W.-J.; Azhar, S.; Kraemer, F.B. Lipid droplets and steroidogenic cells. Exp. Cell Res. 2016, 340, 209–214. [Google Scholar] [CrossRef] [Green Version]
  3. Narayan, P.; Ulloa-Aguirre, A.; Dias, J.A. Chapter 2—Gonadotropin hormones and their receptors. In Yen & Jaffe’s Reproductive Endocrinology–Physiology, Pathophysiology, and Clinical Management, 8th ed.; Strauss, J.F., III, Barbieri, R.L., Eds.; Elsevier: Philadelphia, PA, USA, 2019; pp. 25–57.e15. ISBN 978-0-323-47912-7. [Google Scholar]
  4. Dufau, M.L. The luteinizing hormone receptor. Annu. Rev. Physiol. 1998, 60, 461–496. [Google Scholar] [CrossRef] [PubMed]
  5. Cattanach, B.M.; Iddon, C.A.; Charlton, H.M.; Chiappa, S.A.; Fink, G. Gonadotrophin-releasing hormone deficiency in a mutant mouse with hypogonadism. Nature 1977, 269, 338–340. [Google Scholar] [CrossRef] [PubMed]
  6. Baker, P.J.; O’Shaughnessy, P.J. Role of gonadotrophins in regulating numbers of Leydig and Sertoli cells during fetal and postnatal development in mice. Reproduction 2001, 122, 227–234. [Google Scholar] [CrossRef] [PubMed]
  7. Lei, Z.M.; Mishra, S.; Zou, W.; Xu, B.; Foltz, M.; Li, X.; Rao, C.V. Targeted disruption of luteinizing hormone/human chorionic gonadotropin receptor gene. Mol. Endocrinol. 2001, 15, 184–200. [Google Scholar] [CrossRef]
  8. Zhang, F.P.; Poutanen, M.; Wilbertz, J.; Huhtaniemi, I. Normal prenatal but arrested postnatal sexual development of luteinizing hormone receptor knockout (LuRKO) mice. Mol. Endocrinol. 2001, 15, 172–183. [Google Scholar] [CrossRef]
  9. Ma, X.; Dong, Y.; Matzuk, M.M.; Kumar, T.R. Targeted disruption of luteinizing hormone β-subunit leads to hypogonadism, defects in gonadal steroidogenesis, and infertility. Proc. Natl. Acad. Sci. USA 2004, 101, 17294–17299. [Google Scholar] [CrossRef]
  10. Pakarainen, T.; Zhang, F.P.; Mäkelä, S.; Poutanen, M.; Huhtaniemi, I. Testosterone replacement therapy induces spermatogenesis and partially restores fertility in luteinizing hormone receptor knockout mice. Endocrinology 2005, 146, 596–606. [Google Scholar] [CrossRef] [Green Version]
  11. Narayan, P. Genetic Models for the Study of Luteinizing Hormone Receptor Function. Front. Endocrinol. 2015, 6, 152. [Google Scholar] [CrossRef] [Green Version]
  12. Latronico, A.; Arnhold, I.P. Inactivating mutations of the human luteinizing hormone receptor in both sexes. Semin. Reprod. Med. 2012, 30, 382–386. [Google Scholar] [CrossRef] [PubMed]
  13. Clayton, R.N. Gonadotrophin receptors. Baillière’s Clin. Endocrinol. Metab. 1996, 10, 1–8. [Google Scholar] [CrossRef] [PubMed]
  14. Hofmann, F.; Bechtel, P.J.; Krebs, E.G. Concentrations of cyclic AMP dependent protein kinase subunits in various tissues. J. Biol. Chem. 1977, 252, 1441–1447. [Google Scholar] [CrossRef] [PubMed]
  15. Taylor, S.S.; Zheng, J.; Radzio-Andzelm, E.; Knighton, D.R.; Ten Eyck, L.F.; Sowadski, J.M.; Herberg, F.W.; Yonemoto, W.M. cAMP-dependent protein kinase defines a family of enzymes. Philos. Trans. R. Soc. Lond. B Biol. Sci. 1993, 340, 315–324. [Google Scholar] [CrossRef]
  16. Walsh, D.A.; Perkins, J.P.; Krebs, E.G. An adenosine 3′,5′-monophosphate-dependant protein kinase from rabbit skeletal muscle. J. Biol. Chem. 1968, 243, 3763–3765. [Google Scholar] [CrossRef] [PubMed]
  17. Kim, H.A.; DeClue, J.E.; Ratner, N. cAMP-dependent protein kinase a is required for Schwann cell growth: Interactions between the cAMP and neuregulin/tyrosine kinase pathways. J. Neurosci. Res. 1997, 49, 236–247. [Google Scholar] [CrossRef]
  18. London, E.; Stratakis, C.A. The regulation of PKA signaling in obesity and in the maintenance of metabolic health. Pharmacol. Ther. 2022, 237, 108113. [Google Scholar] [CrossRef]
  19. Keryer, G.; Alsat, E.; Taskén, K.; Evain-Brion, D. Cyclic AMP-dependent protein kinases and human trophoblast cell differentiation in vitro. J. Cell Sci. 1998, 111, 995–1004. [Google Scholar] [CrossRef]
  20. Amato, S.F.; Nakajima, K.; Hirano, T.; Chiles, T.C. Transcriptional regulation of the junB gene in B lymphocytes: Role of protein kinase A and a membrane Ig-regulated protein phosphatase. J. Immunol. 1997, 159, 4676–4685. [Google Scholar] [CrossRef]
  21. Pariset, C.; Feinberg, J.; Dacheux, J.L.; Oyen, O.; Jahnsen, T.; Weinman, S. Differential expression and subcellular localization for subunits of cAMP-dependent protein kinase during ram spermatogenesis. J. Cell Biol. 1989, 109, 1195–1205. [Google Scholar] [CrossRef]
  22. Baum, B.J.; Colpo, F.T.; Filburn, C.R. Characterization and relationship to exocrine secretion of rat parotid gland cyclic AMP-dependent protein kinase. Arch. Oral Biol. 1981, 26, 333–337. [Google Scholar] [CrossRef] [PubMed]
  23. Hincke, M.T.; Soor, S.K. Stimulation of rat parotid secretion by cAMP analogues that synergistically activate the type II isoenzyme of the cAMP-dependent protein kinase. Arch. Oral Biol. 1992, 37, 85–92. [Google Scholar] [CrossRef] [PubMed]
  24. Cadd, G.; McKnight, G.S. Distinct patterns of cAMP-dependent protein kinase gene expression in mouse brain. Neuron 1989, 3, 71–79. [Google Scholar] [CrossRef] [PubMed]
  25. Skålhegg, B.S.; Huang, Y.; Su, T.; Idzerda, R.L.; Stanley McKnight, G.; Burton, K.A. Mutation of the Cα subunit of PKA leads to growth retardation and sperm dysfunction. Mol. Endocrinol. 2002, 16, 630–639. [Google Scholar] [CrossRef] [Green Version]
  26. Veugelers, M.; Wilkes, D.; Burton, K.; McDermott, D.A.; Song, Y.; Goldstein, M.M.; La Perle, K.; Vaughan, C.J.; O’Hagan, A.; Bennett, K.R.; et al. Comparative PRKAR1A genotype–phenotype analyses in humans with Carney complex and prkar1a haploinsufficient mice. Proc. Natl. Acad. Sci. USA 2004, 101, 14222–14227. [Google Scholar] [CrossRef]
  27. Stratakis, C.A. Mutations of the gene encoding the protein kinase A type I-alpha regulatory subunit (PRKAR1A) in patients with the “complex of spotty skin pigmentation, myxomas, endocrine overactivity, and schwannomas” (Carney complex). Ann. N. Y. Acad. Sci. 2002, 968, 3–21. [Google Scholar] [CrossRef]
  28. Tremblay, J.J.; Viger, R.S. Transcription factor GATA-4 is activated by phosphorylation of serine 261 via the cAMP/protein kinase A signaling pathway in gonadal cells. J. Biol. Chem. 2003, 278, 22128–22135. [Google Scholar] [CrossRef] [Green Version]
  29. Pierre, K.J.; Tremblay, J.J. Differential response of transcription factors to activated kinases in steroidogenic and non-steroidogenic cells. Int. J. Mol. Sci. 2022, 23, 13153. [Google Scholar] [CrossRef]
  30. Costa, R.R.; Varanda, W.A.; Franci, C.R. A calcium-induced calcium release mechanism supports luteinizing hormone-induced testosterone secretion in mouse Leydig cells. Am. J. Physiol. Cell Physiol. 2010, 299, 316–323. [Google Scholar] [CrossRef] [Green Version]
  31. Patergnani, S.; Danese, A.; Bouhamida, E.; Aguiari, G.; Previati, M.; Pinton, P.; Giorgi, C. Various aspects of calcium signaling in the regulation of apoptosis, autophagy, cell proliferation, and cancer. Int. J. Mol. Sci. 2020, 21, 8323. [Google Scholar] [CrossRef]
  32. Koch, G.L. The endoplasmic reticulum and calcium storage. Bioessays 1990, 12, 527–531. [Google Scholar] [CrossRef] [PubMed]
  33. Berridge, M.J.; Lipp, P.; Bootman, M.D. The versatility and universality of calcium signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef] [PubMed]
  34. Sullivan, M.H.F.; Cooke, B.A. The role of Ca2+ in steroidogenesis in Leydig cells. Biochem. J. 1986, 236, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Sullivan, M.H.; Cooke, B.A. The effect of calcium on the potentiation of LH-stimulated steroidogenesis and inhibition of LH-stimulated cyclic AMP production by LHRH agonist (ICI 118630) in rat Leydig cells. Mol. Cell. Endocrinol. 1984, 34, 17–22. [Google Scholar] [CrossRef] [PubMed]
  36. Abdou, H.S.; Villeneuve, G.; Tremblay, J.J. The calcium signaling pathway regulates leydig cell steroidogenesis through a transcriptional cascade involving the nuclear receptor NR4A1 and the steroidogenic acute regulatory protein. Endocrinology 2013, 154, 511–520. [Google Scholar] [CrossRef] [Green Version]
  37. Beghi, S.; Furmanik, M.; Jaminon, A.; Veltrop, R.; Rapp, N.; Wichapong, K.; Bidar, E.; Buschini, A.; Schurgers, L.J. Calcium signalling in heart and vessels: Role of calmodulin and downstream calmodulin-pependent protein kinases. Int. J. Mol. Sci. 2022, 23, 16139. [Google Scholar] [CrossRef]
  38. Martin, L.J.; Boucher, N.; Brousseau, C.; Tremblay, J.J. The orphan nuclear receptor NUR77 regulates hormone-induced StAR transcription in Leydig cells through cooperation with Ca2+/calmodulin-dependent protein kinase I. Mol. Endocrinol. 2008, 22, 2021–2037. [Google Scholar] [CrossRef] [Green Version]
  39. Lee, J.C.; Kwon, Y.G.; Lawrence, D.S.; Edelman, A.M. A requirement of hydrophobic and basic amino acid residues for substrate recognition by Ca2+/calmodulin-dependent protein kinase Ia. Proc. Natl. Acad. Sci. USA 1994, 91, 6413–6417. [Google Scholar] [CrossRef]
  40. Picciotto, M.R.; Nastiuk, K.L.; Nairn, A.C. Structure, regulation, and function of calcium/calmodulin-dependent protein kinase I. Adv. Pharmacol. 1996, 36, 251–275. [Google Scholar] [CrossRef]
  41. Haribabu, B.; Hook, S.S.; Selbert, M.A.; Goldstein, E.G.; Tomhave, E.D.; Edelman, A.M.; Snyderman, R.; Means, A.R. Human calcium-calmodulin dependent protein kinase I: cDNA cloning, domain structure and activation by phosphorylation at threonine-177 by calcium-calmodulin dependent protein kinase I kinase. EMBO J. 1995, 14, 3679–3686. [Google Scholar] [CrossRef]
  42. Tremblay, J.J. Molecular regulation of steroidogenesis in endocrine Leydig cells. Steroids 2015, 103, 3–10. [Google Scholar] [CrossRef] [PubMed]
  43. de Mattos, K.; Viger, R.S.; Tremblay, J.J. Transcription factors in the regulation of Leydig cell gene expression and function. Front. Endocrinol. 2022, 13, 881309. [Google Scholar] [CrossRef] [PubMed]
  44. Morris, J.K.; Richards, J.S. Luteinizing hormone induces prostaglandin endoperoxide synthase-2 and luteinization in vitro by A-kinase and C-kinase pathways. Endocrinology 1995, 136, 1549–1558. [Google Scholar] [CrossRef] [PubMed]
  45. Clemens, M.J.; Trayner, I.; Menaya, J. The role of protein kinase C isoenzymes in the regulation of cell proliferation and differentiation. J. Cell Sci. 1992, 103, 881–887. [Google Scholar] [CrossRef]
  46. Chen, J.-L.; Lin, H.H.; Kim, K.-J.; Lin, A.; Ou, J.-H.J.; Ann, D.K. PKC delta signaling: A dual role in regulating hypoxic stress-induced autophagy and apoptosis. Autophagy 2009, 5, 244–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Gerbino, A.; De Zio, R.; Russo, D.; Milella, L.; Milano, S.; Procino, G.; Pusch, M.; Svelto, M.; Carmosino, M. Role of PKC in the regulation of the human kidney chloride channel ClC-Ka. Sci. Rep. 2020, 10, 10268. [Google Scholar] [CrossRef]
  48. Garcia-Concejo, A.; Larhammar, D. Protein kinase C family evolution in jawed vertebrates. Dev. Biol. 2021, 479, 77–90. [Google Scholar] [CrossRef]
  49. Kawano, T.; Inokuchi, J.; Eto, M.; Murata, M.; Kang, J.H. Activators and inhibitors of protein kinase c (Pkc): Their applications in clinical trials. Pharmaceutics 2021, 13, 1748. [Google Scholar] [CrossRef]
  50. Kang, J.H.; Toita, R.; Kim, C.W.; Katayama, Y. Protein kinase C (PKC) isozyme-specific substrates and their design. Biotechnol. Adv. 2012, 30, 1662–1672. [Google Scholar] [CrossRef]
  51. Manna, P.R.; Soh, J.W.; Stocco, D.M. The involvement of specific PKC isoenzymes in phorbol ester-mediated regulation of steroidogenic acute regulatory protein expression and steroid synthesis in mouse leydig cells. Endocrinology 2011, 152, 313–325. [Google Scholar] [CrossRef] [Green Version]
  52. Pelosin, J.M.; Ricouart, A.; Sergheraert, C.; Benahmed, M.; Chambaz, E.M. Expression of protein kinase C isoforms in various steroidogenic cell types. Mol. Cell. Endocrinol. 1991, 75, 149–155. [Google Scholar] [CrossRef]
  53. Manna, P.R.; Huhtaniemi, I.T.; Stocco, D.M. Mechanisms of protein kinase C signaling in the modulation of 3′,5′-cyclic adenosine monophosphate-mediated steroidogenesis in mouse gonadal cells. Endocrinology 2009, 150, 3308–3317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Jo, Y.; King, S.R.; Khan, S.A.; Stocco, D.M. Involvement of protein kinase C and cyclic adenosine 3′,5′-monophosphate-dependent kinase in steroidogenic acute regulatory protein expression and steroid biosynthesis in Leydig cells. Biol. Reprod. 2005, 73, 244–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Manna, P.R.; Jo, Y.; Stocco, D.M. Regulation of Leydig cell steroidogenesis by extracellular signal-regulated kinase 1/2: Role of protein kinase A and protein kinase C signaling. J. Endocrinol. 2007, 193, 53–63. [Google Scholar] [CrossRef] [PubMed]
  56. Esteban-Villarrubia, J.; Soto-Castillo, J.J.; Pozas, J.; Román-Gil, M.S.; Orejana-Martín, I.; Torres-Jiménez, J.; Carrato, A.; Alonso-Gordoa, T.; Molina-Cerrillo, J. Tyrosine kinase receptors in oncology. Int. J. Mol. Sci. 2020, 21, 8529. [Google Scholar] [CrossRef]
  57. Roux, P.P.; Blenis, J. ERK and p38 MAPK-activated protein kinases: A family of protein kinases with diverse biological functions. Microbiol. Mol. Biol. Rev. 2004, 68, 320–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Roskoski, R. ERK1/2 MAP kinases: Structure, function, and regulation. Pharmacol. Res. 2012, 66, 105–143. [Google Scholar] [CrossRef]
  59. Plotnikov, A.; Zehorai, E.; Procaccia, S.; Seger, R. The MAPK cascades: Signaling components, nuclear roles and mechanisms of nuclear translocation. Biochim. Biophys. Acta-Mol. Cell Res. 2011, 1813, 1619–1633. [Google Scholar] [CrossRef] [Green Version]
  60. Shiraishi, K.; Ascoli, M. Lutropin/choriogonadotropin stimulate the proliferation of primary cultures of rat leydig cells through a pathway that involves activation of the extracellularly regulated kinase 1/2 cascade. Endocrinology 2007, 148, 3214–3225. [Google Scholar] [CrossRef] [Green Version]
  61. Yamashita, S.; Tai, P.; Charron, J.; Ko, C.M.; Ascoli, M. The leydig cell MEK/ERK pathway is critical for maintaining a functional population of adult leydig cells and for fertility. Mol. Endocrinol. 2011, 25, 1211–1222. [Google Scholar] [CrossRef] [Green Version]
  62. Manna, P.R.; Stocco, D.M. The role of specific mitogen-activated protein kinase signaling cascades in the regulation of steroidogenesis. J. Signal Transduct. 2011, 2011, 821615. [Google Scholar] [CrossRef] [Green Version]
  63. Matzkin, M.E.; Yamashita, S.; Ascoli, M. The ERK1/2 pathway regulates testosterone synthesis by coordinately regulating the expression of steroidogenic genes in Leydig cells. Mol. Cell. Endocrinol. 2013, 370, 130–137. [Google Scholar] [CrossRef] [Green Version]
  64. Hirakawa, T.; Ascoli, M. The lutropin/choriogonadotropin receptor-induced phosphorylation of the extracellular signal-regulated kinases in Leydig cells is mediated by a protein kinase A-dependent activation of ras. Mol. Endocrinol. 2003, 17, 2189–2200. [Google Scholar] [CrossRef]
  65. Martinat, N.; Crépieux, P.; Reiter, E.; Guillou, F. Extracellular signal-regulated kinases (ERK) 1,2 are required for luteinizing hormone (LH)-induced steroidogenesis in primary Leydig cells and control steroidogenic acute regulatory (StAR) expression. Reprod. Nutr. Dev. 2005, 45, 101–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Liang, Q.; Wiese, R.J.; Bueno, O.F.; Dai, Y.-S.; Markham, B.E.; Molkentin, J.D. The transcription factor GATA4 is activated by extracellular signal-regulated kinase 1- and 2-mediated phosphorylation of serine 105 in cardiomyocytes. Mol. Cell. Biol. 2001, 21, 7460–7469. [Google Scholar] [CrossRef] [Green Version]
  67. Bergeron, F.; Boulende Sab, A.; Bouchard, M.F.; Taniguchi, H.; Souchkova, O.; Brousseau, C.; Tremblay, J.J.; Pilon, N.; Viger, R.S. Phosphorylation of GATA4 serine 105 but not serine 261 is required for testosterone production in the male mouse. Andrology 2019, 7, 357–372. [Google Scholar] [CrossRef]
  68. Coffer, P.J.; Woodgett, J.R. Molecular cloning and characterisation of a novel putative protein-serine kinase related to the cAMP-dependent and protein kinase C families. Eur. J. Biochem. 1991, 201, 475–481. [Google Scholar] [CrossRef] [PubMed]
  69. Jones, P.F.; Jakubowicz, T.; Hemmings, B.A. Molecular cloning of a second form of rac protein kinase. Mol. Biol. Cell 1991, 2, 1001–1009. [Google Scholar] [CrossRef] [Green Version]
  70. Jones, P.F.; Jakubowicz, T.; Pitossi, F.J.; Maurer, F.; Hemmings, B.A. Molecular cloning and identification of a serine/threonine protein kinase of the second-messenger subfamily. Proc. Natl. Acad. Sci. USA 1991, 88, 4171–4175. [Google Scholar] [CrossRef]
  71. Bellacosa, A.; Testa, J.R.; Staal, S.P.; Tsichlis, P.N. A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science 1991, 254, 274–277. [Google Scholar] [CrossRef] [PubMed]
  72. Manning, B.D.; Cantley, L.C. AKT/PKB signaling: Navigating downstream. Cell 2007, 129, 1261–1274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Alessi, D.R.; Andjelkovic, M.; Caudwell, B.; Cron, P.; Morrice, N.; Cohen, P.; Hemmings, B.A. Mechanism of activation of protein kinase B by insulin and IGF-1. EMBO J. 1996, 15, 6541–6551. [Google Scholar] [CrossRef]
  74. Li, W.; Amri, H.; Huang, H.; Wu, C.; Papadopoulos, V. Gene and protein profiling of the response of MA-10 Leydig tumor cells to human chorionic gonadotropin. J. Androl. 2004, 25, 900–913. [Google Scholar] [CrossRef] [PubMed]
  75. Chesnokova, V.; Melmed, S. Non-pituitary GH regulation of the tissue microenvironment. Endocr. Relat. Cancer 2023, 30, e230028. [Google Scholar] [CrossRef]
  76. Bramley, T.A.; Menzies, G.S.; McNeilly, A.S.; Friesen, H.G. Receptors for lactogenic hormones in the ovine corpus luteum. I: A major discrepancy in the specific binding of radiolabelled ovine prolactin and human growth hormone. J. Endocrinol. 1987, 113, 365–374. [Google Scholar] [CrossRef]
  77. Bresson, J.L.; Jeay, S.; Gagnerault, M.C.; Kayser, C.; Beressi, N.; Wu, Z.; Kinet, S.; Dardenne, M.; Postel-Vinay, M.C. Growth hormone (GH) and prolactin receptors in human peripheral blood mononuclear cells: Relation with age and GH-binding protein. Endocrinology 1999, 140, 3203–3209. [Google Scholar] [CrossRef]
  78. Ipsa, E.; Cruzat, V.F.; Kagize, J.N.; Yovich, J.L.; Keane, K.N. Growth hormone and insulin-like growth factor action in reproductive tissues. Front. Endocrinol. 2019, 10, 777. [Google Scholar] [CrossRef] [PubMed]
  79. Dosouto, C.; Calaf, J.; Polo, A.; Haahr, T.; Humaidan, P. Growth hormone and reproduction: Lessons learned from animal models and clinical trials. Front. Endocrinol. 2019, 10, 404. [Google Scholar] [CrossRef]
  80. Lobie, P.E.; Breipohl, W.; Aragón, J.G.; Waters, M.J. Cellular localization of the growth hormone receptor/ binding protein in the male and female reproductive systems. Endocrinology 1990, 126, 2214–2221. [Google Scholar] [CrossRef]
  81. Hull, K.L.; Harvey, S. Growth hormone and reproduction: A review of endocrine and autocrine/paracrine interactions. Int. J. Endocrinol. 2014, 2014, 234014. [Google Scholar] [CrossRef]
  82. Chandrashekar, V.; Bartke, A.; Coschigano, K.T.; Kopchick, J.J. Pituitary and testicular function in growth hormone receptor gene knockout mice. Endocrinology 1999, 140, 1082–1088. [Google Scholar] [CrossRef]
  83. Chandrashekar, V.; Bartke, A.; Awoniyi, C.A.; Tsai-Morris, C.H.; Dufau, M.L.; Russell, L.D.; Kopchick, J.J. Testicular endocrine function in GH receptor gene disrupted mice. Endocrinology 2001, 142, 3443–3450. [Google Scholar] [CrossRef]
  84. Keene, D.E.; Suescun, M.O.; Bostwick, M.G.; Chandrashekar, V.; Bartke, A.; Kopchick, J.J. Puberty is delayed in male growth hormone receptor gene-disrupted mice. J. Androl. 2002, 23, 661–668. [Google Scholar]
  85. Bartlett, J.M.; Charlton, H.M.; Robinson, I.C.; Nieschlag, E. Pubertal development and testicular function in the male growth hormone-deficient rat. J. Endocrinol. 1990, 126, 193–201. [Google Scholar] [CrossRef]
  86. Maran, R.R.M.; Sivakumar, R.; Ravisankar, B.; Valli, G.; Ravichandran, K.; Arunakaran, J.; Aruldhas, M.M. Growth hormone directly stimulates testosterone and oestradiol secretion by rat Leydig cells in vitro and modulates the effects of LH and T3. Endocr. J. 2000, 47, 111–118. [Google Scholar] [CrossRef] [Green Version]
  87. Hébert-Mercier, P.O.; Bergeron, F.; Robert, N.M.; Mehanovic, S.; Pierre, K.J.; Mendoza-Villarroel, R.E.; de Mattos, K.; Brousseau, C.; Tremblay, J.J. Growth hormone-induced STAT5B regulates star gene expression through a cooperation with cJUN in mouse MA-10 Leydig cells. Endocrinology 2022, 163, bqab267. [Google Scholar] [CrossRef] [PubMed]
  88. Kanzaki, M.; Morris, P.L. Growth hormone regulates steroidogenic acute regulatory protein expression and steroidogenesis in Leydig cell progenitors. Endocrinology 1999, 140, 1681–1686. [Google Scholar] [CrossRef]
  89. Spiteri-Grech, J.; Nieschlag, E. The role of growth hormone and insulin-like growth factor I in the regulation of male reproductive function. Horm. Res. 1992, 38 (Suppl. S1), 22–27. [Google Scholar] [CrossRef] [PubMed]
  90. Huh, K.; Nah, W.H.; Xu, Y.; Park, M.J.; Gye, M.C. Effects of recombinant human growth hormone on the onset of puberty, leydig cell differentiation, spermatogenesis and hypothalamic kiss1 expression in immature male rats. World J. Mens Health 2021, 38, 381–388. [Google Scholar] [CrossRef]
  91. Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef] [PubMed]
  92. Rane, S.G.; Reddy, E.P. Janus kinases: Components of multiple signaling pathways. Oncogene 2000, 19, 5662–5679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Parganas, E.; Wang, D.; Stravopodis, D.; Topham, D.J.; Marine, J.C.; Teglund, S.; Vanin, E.F.; Bodner, S.; Colamonici, O.R.; van Deursen, J.M.; et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998, 93, 385–395. [Google Scholar] [CrossRef] [Green Version]
  94. Rodig, S.J.; Meraz, M.A.; White, J.M.; Lampe, P.A.; Riley, J.K.; Arthur, C.D.; King, K.L.; Sheehan, K.C.; Yin, L.; Pennica, D.; et al. Disruption of the Jak1 gene demonstrates obligatory and nonredundant roles of the Jaks in cytokine-induced biologic responses. Cell 1998, 93, 373–383. [Google Scholar] [CrossRef] [Green Version]
  95. Neubauer, H.; Cumano, A.; Müller, M.; Wu, H.; Huffstadt, U.; Pfeffer, K. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell 1998, 93, 397–409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Dehkhoda, F.; Lee, C.M.M.; Medina, J.; Brooks, A.J. The growth hormone receptor: Mechanism of receptor activation, cell signaling, and physiological aspects. Front. Endocrinol. 2018, 9, 35. [Google Scholar] [CrossRef] [Green Version]
  97. Kanzaki, M.; Morris, P.L. Lactogenic hormone-inducible phosphorylation and gamma-activated site-binding activities of Stat5b in primary rat Leydig cells and MA-10 mouse Leydig tumor cells. Endocrinology 1998, 139, 1872–1882. [Google Scholar] [CrossRef]
  98. Chapin, R.E.; Ball, D.J.; Radi, Z.A.; Kumpf, S.W.; Koza-Taylor, P.H.; Potter, D.M.; Mark Vogel, W. Effects of the Janus kinase inhibitor, tofacitinib, on testicular Leydig cell hyperplasia and adenoma in rats, and on prolactin signaling in cultured primary rat Leydig cells. Toxicol. Sci. 2017, 155, 148–156. [Google Scholar] [CrossRef]
  99. Ocampo Daza, D.; Larhammar, D. Evolution of the growth hormone, prolactin, prolactin 2 and somatolactin family. Gen. Comp. Endocrinol. 2018, 264, 94–112. [Google Scholar] [CrossRef]
  100. Raut, S.; Deshpande, S.; Balasinor, N.H. Unveiling the role of prolactin and its receptor in male reproduction. Horm. Metab. Res. 2019, 51, 215–219. [Google Scholar] [CrossRef] [Green Version]
  101. Bex, F.J.; Bartke, A. Testicular LH binding in the hamster: Modification by photoperiod and prolactin. Endocrinology 1977, 100, 1223–1226. [Google Scholar] [CrossRef]
  102. Takase, M.; Tsutsui, K.; Kawashima, S. Effects of PRL and FSH on LH binding and number of Leydig cells in hypophysectomized mice. Endocrinol. Jpn. 1990, 37, 193–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Dombrowicz, D.; Sente, B.; Closset, J.; Hennen, G. Dose-dependent effects of human prolactin on the immature hypophysectomized rat testis. Endocrinology 1992, 130, 695–700. [Google Scholar] [CrossRef] [PubMed]
  104. Purvis, K.; Clausen, O.P.F.; Olsen, A.; Haug, E.; Hansson, V. Prolactin and Leydig cell responsiveness to LH/hCG in the rat. Syst. Biol. Reprod. Med. 1979, 3, 219–230. [Google Scholar] [CrossRef] [PubMed]
  105. Klemcke, H.G.; Bartke, A.; Borer, K.T. Regulation of testicular prolactin and luteinizing hormone receptors in golden hamsters. Endocrinology 1984, 114, 594–603. [Google Scholar] [CrossRef]
  106. Aragona, C.; Bohnet, H.G.; Friesen, H.G. Localization of prolactin binding in prostate and testis: The role of serum prolactin concentration on the testicular LH receptor. Acta Endocrinol. 1977, 84, 402–409. [Google Scholar] [CrossRef]
  107. Grizard, G.; Andre, M.; Jarrige, J.F.; Chambon, M.; Boucher, D. Effects of bromocriptine on pituitary-testicular function in the rat: Possible inhibition of in vitro production of androgen by Leydig cells. Int. J. Androl. 1983, 6, 563–574. [Google Scholar] [CrossRef] [PubMed]
  108. Waeber, C.; Reymond, O.; Reymond, M.; Lemarchand-Beraud, T. Effects of hyper- and hypoprolactinemia on gonadotropin secretion, rat testicular luteinizing hormone/human chorionic gonadotropin receptors and testosterone production by isolated Leydig cells. Biol. Reprod. 1983, 28, 167–177. [Google Scholar] [CrossRef] [Green Version]
  109. Weiss-Messer, E.; Ber, R.; Barkey, R.J. Prolactin and MA-10 Leydig cell steroidogenesis: Biphasic effects of prolactin and signal transduction. Endocrinology 1996, 137, 5509–5518. [Google Scholar] [CrossRef]
  110. Steger, R.W.; Chandrashekar, V.; Zhao, W.; Bartke, A.; Horseman, N.D. Neuroendocrine and reproductive functions in male mice with targeted disruption of the prolactin gene. Endocrinology 1998, 139, 3691–3695. [Google Scholar] [CrossRef]
  111. Binart, N.; Melaine, N.; Pineau, C.; Kercret, H.; Touzalin, A.M.; Imbert-Bolloré, P.; Kelly, P.A.; Jégou, B. Male reproductive function is not affected in prolactin receptor-deficient mice. Endocrinology 2003, 144, 3779–3782. [Google Scholar] [CrossRef] [Green Version]
  112. Ji, M.; Chen, D.; Zhao, X.; Huang, F.; Guan, X.; Wen, X.; Wang, J.; Shao, J.; Xie, J.; Shan, D.; et al. Isolation of Leydig cells from adult rat testes by magnetic-activated cell sorting protocol based on prolactin receptor expression. Andrology 2022, 10, 1197–1207. [Google Scholar] [CrossRef] [PubMed]
  113. Jabbour, H.N.; Lincoln, G.A. Prolactin receptor expression in the testis of the ram: Localisation, functional activation and the influence of gonadotrophins. Mol. Cell. Endocrinol. 1999, 148, 151–161. [Google Scholar] [CrossRef] [PubMed]
  114. Hair, W.M.; Gubbay, O.; Jabbour, H.N.; Lincoln, G.A. Prolactin receptor expression in human testis and accessory tissues: Localization and function. Mol. Hum. Reprod. 2002, 8, 606–611. [Google Scholar] [CrossRef] [PubMed]
  115. Barkey, R.J.; Weiss-Messer, E.; Hacham, H.; Herscovich, S.; Ber, R.; Amit, T. Prolactin and testicular Leydig cell function: Characterization of prolactin receptors in the murine MA-10 testicular Leydig cell line. Proc. Soc. Exp. Biol. Med. 1994, 206, 243–248. [Google Scholar] [CrossRef]
  116. Wahlström, T.; Huhtaniemi, I.; Hovatta, O.; Seppälä, M. Localization of luteinizing hormone, follicle-stimulating hormone, prolactin, and their receptors in human and rat testis using immunohistochemistry and radioreceptor assay. J. Clin. Endocrinol. Metab. 1983, 57, 825–830. [Google Scholar] [CrossRef]
  117. Bole-Feysot, C.; Goffin, V.; Edery, M.; Binart, N.; Kelly, P.A. Prolactin (PRL) and its receptor: Actions, signal transduction pathways and phenotypes observed in PRL receptor knockout mice. Endocr. Rev. 1998, 19, 225–268. [Google Scholar] [CrossRef]
  118. Manna, P.R.; El-Hefnawy, T.; Kero, J.; Huhtaniemi, I.T. Biphasic action of prolactin in the regulation of murine Leydig tumor cell functions. Endocrinology 2001, 142, 308–318. [Google Scholar] [CrossRef]
  119. Griffeth, R.J.; Bianda, V.; Nef, S. The emerging role of insulin-like growth factors in testis development and function. Basic Clin. Androl. 2014, 24, 12. [Google Scholar] [CrossRef]
  120. Ashcroft, F.M.; Proks, P.; Smith, P.A.; Ammälä, C.; Bokvist, K.; Rorsman, P. Stimulus-secretion coupling in pancreatic beta cells. J. Cell. Biochem. 1994, 55, 54–65. [Google Scholar] [CrossRef]
  121. Röder, P.V.; Wu, B.; Liu, Y.; Han, W. Pancreatic regulation of glucose homeostasis. Exp. Mol. Med. 2016, 48, e219. [Google Scholar] [CrossRef] [Green Version]
  122. Maresch, C.C.; Stute, D.C.; Alves, M.G.; Oliveira, P.F.; de Kretser, D.M.; Linn, T. Diabetes-induced hyperglycemia impairs male reproductive function: A systematic review. Hum. Reprod. Update 2018, 24, 86–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Kitamura, T.; Kahn, C.R.; Accili, D. Insulin receptor knockout mice. Annu. Rev. Physiol. 2003, 65, 313–332. [Google Scholar] [CrossRef]
  124. Ballester, J.; Muñoz, M.C.; Domínguez, J.; Rigau, T.; Guinovart, J.J.; Rodríguez-Gil, J.E. Insulin-dependent diabetes affects testicular function by FSH- and LH-linked mechanisms. J. Androl. 2004, 25, 706–719. [Google Scholar] [CrossRef] [PubMed]
  125. Schoeller, E.L.; Albanna, G.; Frolova, A.I.; Moley, K.H. Insulin rescues impaired spermatogenesis via the hypothalamic-pituitary- gonadal axis in Akita diabetic mice and restores male fertility. Diabetes 2012, 61, 1869–1878. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Wagner, I.V.; Klöting, N.; Savchuk, I.; Eifler, L.; Kulle, A.; Kralisch-Jäcklein, S.; Dötsch, J.; Hiort, O.; Svechnikov, K.; Söder, O. Diabetes Type 1 Negatively Influences Leydig Cell Function in Rats, Which is Partially Reversible by Insulin Treatment. Endocrinol. 2021, 162, bqab017. [Google Scholar] [CrossRef]
  127. Navarro-Casado, L.; Juncos-Tobarra, M.A.; Cháfer-Rudilla, M.; Íñiguez De Onzoño, L.; Blázquez-Cabrera, J.A.; Miralles-García, J.M. Effect of experimental diabetes and STZ on male fertility capacity. Study in rats. J. Androl. 2010, 31, 584–592. [Google Scholar] [CrossRef]
  128. Ermetici, F.; Donadio, F.; Iorio, L.; Malavazos, A.E.; Dolci, A.; Peverelli, E.; Barbieri, A.M.; Morricone, L.; Chiodini, I.; Arosio, M.; et al. Peripheral insulin-like factor 3 concentrations are reduced in men with type 2 diabetes mellitus: Effect of glycemic control and visceral adiposity on Leydig cell function. Eur. J. Endocrinol. 2009, 161, 853–859. [Google Scholar] [CrossRef] [Green Version]
  129. Neirijnck, Y.; Calvel, P.; Kilcoyne, K.R.; Kühne, F.; Stévant, I.; Griffeth, R.J.; Pitetti, J.L.; Andric, S.A.; Hu, M.C.; Pralong, F.; et al. Insulin and IGF1 receptors are essential for the development and steroidogenic function of adult Leydig cells. FASEB J. 2018, 32, 3321–3335. [Google Scholar] [CrossRef] [Green Version]
  130. Lin, T.; Haskell, J.; Vinson, N.; Terracio, L. Characterization of insulin and insulin-like growth factor i receptors of purified leydig cells and their role in steroidogenesis in primary culture: A comparative study. Endocrinology 1986, 119, 1641–1647. [Google Scholar] [CrossRef]
  131. Leisegang, K.; Henkel, R. The in vitro modulation of steroidogenesis by inflammatory cytokines and insulin in TM3 Leydig cells. Reprod. Biol. Endocrinol. 2018, 16, 26. [Google Scholar] [CrossRef] [Green Version]
  132. Ahn, S.W.; Gang, G.T.; Kim, Y.D.; Ahn, R.S.; Harris, R.A.; Lee, C.H.; Choi, H.S. Insulin directly regulates steroidogenesis via induction of the orphan nuclear receptor DAX-1 in testicular Leydig cells. J. Biol. Chem. 2013, 288, 15937–15946. [Google Scholar] [CrossRef] [Green Version]
  133. Avruch, J. MAP kinase pathways: The first twenty years. Biochim. Biophys. Acta-Mol. Cell Res. 2007, 1773, 1150–1160. [Google Scholar] [CrossRef] [Green Version]
  134. Cannarella, R.; Condorelli, R.A.; La Vignera, S.; Calogero, A.E. Effects of the insulin-like growth factor system on testicular differentiation and function: A review of the literature. Andrology 2018, 6, 3–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Handelsman, D.J.; Spaliviero, J.A.; Scott, C.D.; Baxter, R.C. Identification of insulin-like growth factor-I and its receptors in the rat testis. Acta Endocrinol. 1985, 109, 543–549. [Google Scholar] [CrossRef]
  136. Wang, G.; Hardy, M.P. Development of Leydig cells in the insulin-like growth factor-I (IGF-I) knockout mouse: Effects of IGF-I replacement and gonadotropic stimulation. Biol. Reprod. 2004, 70, 632–639. [Google Scholar] [CrossRef]
  137. Xu, Y.; Han, C.Y.; Park, M.J.; Gye, M.C. Increased testicular insulin-like growth factor 1 is associated with gonadal activation by recombinant growth hormone in immature rats. Reprod. Biol. Endocrinol. 2022, 20, 72. [Google Scholar] [CrossRef] [PubMed]
  138. Cailleau, J.; Vermeire, S.; Verhoeven, G. Independent control of the production of insulin-like growth factor I and its binding protein by cultured testicular cells. Mol. Cell. Endocrinol. 1990, 69, 79–89. [Google Scholar] [CrossRef]
  139. Nagpal, M.L.; Wang, D.; Calkins, J.H.; Chang, W.W.; Lin, T. Human chorionic gonadotropin up-regulates insulin-like growth factor-I receptor Gene expression of Leydig cells. Endocrinology 1991, 129, 2820–2826. [Google Scholar] [CrossRef]
  140. Lin, T.; Blaisdell, J.; Haskell, J.F. Hormonal regulation of type I insulin-like growth factor receptors of Leydig cells in hypophysectomized rats. Endocrinology 1988, 123, 134–139. [Google Scholar] [CrossRef] [PubMed]
  141. Baker, J.; Hardy, M.P.; Zhou, J.; Bondy, C.; Lupu, F.; Bellvé, A.R.; Efstratiadis, A. Effects of an Igf1 gene null mutation on mouse reproduction. Mol. Endocrinol. 1996, 10, 903–918. [Google Scholar] [CrossRef] [Green Version]
  142. Hakuno, F.; Takahashi, S.I. 40 years of IGF1: IGF1 receptor signaling pathways. J. Mol. Endocrinol. 2018, 61, T69–T86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Khan, S.; Teerds, K.; Dorrington, J. Growth factor requirements for DNA synthesis by Leydig cells from the immature rat. Biol. Reprod. 1992, 46, 335–341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Wang, G.M.; O’Shaughnessy, P.J.; Chubb, C.; Robaire, B.; Hardy, M.P. Effects of insulin-like growth factor I on steroidogenic enzyme expression levels in mouse Leydig cells. Endocrinology 2003, 144, 5058–5064. [Google Scholar] [CrossRef] [Green Version]
  145. Moore, A.; Morris, I.D. The involvement of insulin-like growth factor-I in local control of steroidogenesis and DNA synthesis of Leydig and non-Leydig cells in the rat testicular interstitium. J. Endocrinol. 1993, 138, 107–114. [Google Scholar] [CrossRef]
  146. Hu, G.-X.; Lin, H.; Chen, G.-R.; Chen, B.-B.; Lian, Q.-Q.; Hardy, D.O.; Zirkin, B.R.; Ge, R.-S. Deletion of the Igf1 gene: Suppressive effects on adult Leydig cell development. J. Androl. 2010, 31, 379–387. [Google Scholar] [CrossRef] [Green Version]
  147. Colón, E.; Zaman, F.; Axelson, M.; Larsson, O.; Carlsson-Skwirut, C.; Svechnikov, K.V.; Söder, O. Insulin-like growth factor-I is an important antiapoptotic factor for rat Leydig cells during postnatal development. Endocrinology 2007, 148, 128–139. [Google Scholar] [CrossRef]
  148. Tai, P.; Shiraishi, K.; Ascoli, M. Activation of the lutropin/choriogonadotropin receptor inhibits apoptosis of immature Leydig cells in primary culture. Endocrinology 2009, 150, 3766–3773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Villalpando, I.; Lira, E.; Medina, G.; Garcia-Garcia, E.; Echeverria, O. Insulin-like growth factor 1 is expressed in mouse developing testis and regulates somatic cell proliferation. Exp. Biol. Med. 2008, 233, 419–426. [Google Scholar] [CrossRef]
  150. Ornitz, D.M.; Itoh, N. New developments in the biology of fibroblast growth factors. WIREs Mech. Dis. 2022, 14, e1549. [Google Scholar] [CrossRef]
  151. Cotton, L.M.; O’Bryan, M.K.; Hinton, B.T. Cellular signaling by fibroblast growth factors (FGFs) and their receptors (FGFRs) in male reproduction. Endocr. Rev. 2008, 29, 193–216. [Google Scholar] [CrossRef] [Green Version]
  152. Ostrer, H.; Huang, H.Y.; Masch, R.J.; Shapiro, E. A cellular study of human testis development. Sex. Dev. 2007, 1, 286–292. [Google Scholar] [CrossRef] [PubMed]
  153. Lai, M.S.; Wang, C.Y.; Yang, S.H.; Wu, C.C.; Sun, H.S.; Tsai, S.J.; Chuang, J.I.; Chen, Y.C.; Huang, B.M. The expression profiles of fibroblast growth factor 9 and its receptors in developing mice testes. Organogenesis 2016, 12, 61–77. [Google Scholar] [CrossRef] [Green Version]
  154. Gao, X.; Yao, X.; Yang, H.; Deng, K.; Guo, Y.; Zhang, T.; Zhang, G.; Wang, F. Role of FGF9 in sheep testis steroidogenesis during sexual maturation. Anim. Reprod. Sci. 2018, 197, 177–184. [Google Scholar] [CrossRef]
  155. DiNapoli, L.; Batchvarov, J.; Capel, B. FGF9 promotes survival of germ cells in the fetal testis. Development 2006, 133, 1519–1527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Lin, Y.M.; Tsai, C.C.; Chung, C.L.; Chen, P.R.; Sunny Sun, H.; Tsai, S.J.; Huang, B.M. Fibroblast growth factor 9 stimulates steroidogenesis in postnatal Leydig cells. Int. J. Androl. 2010, 33, 545–553. [Google Scholar] [CrossRef]
  157. Colvin, J.S.; Green, R.P.; Schmahl, J.; Capel, B.; Ornitz, D.M. Male-to-female sex reversal in mice lacking fibroblast growth factor 9. Cell 2001, 104, 875–889. [Google Scholar] [CrossRef] [PubMed]
  158. Croft, B.; Bird, A.D.; Ono, M.; Eggers, S.; Bagheri-Fam, S.; Ryan, J.M.; Reyes, A.P.; van den Bergen, J.; Baxendale, A.; Thompson, E.M.; et al. FGF9 variant in 46,XY DSD patient suggests a role for dimerization in sex determination. Clin. Genet. 2023, 103, 277–287. [Google Scholar] [CrossRef]
  159. Lai, M.S.; Cheng, Y.S.; Chen, P.R.; Tsai, S.J.; Huang, B.M. Fibroblast growth factor 9 activates Akt and MAPK pathways to stimulate steroidogenesis in mouse leydig cells. PLoS ONE 2014, 9, e90243. [Google Scholar] [CrossRef] [Green Version]
  160. Zeng, F.; Harris, R.C. Epidermal growth factor, from gene organization to bedside. Semin. Cell Dev. Biol. 2014, 28, 2–11. [Google Scholar] [CrossRef] [Green Version]
  161. Abud, H.E.; Chan, W.H.; Jardé, T. Source and impact of the EGF family of ligands on intestinal stem cells. Front. Cell Dev. Biol. 2021, 9, 685665. [Google Scholar] [CrossRef]
  162. Sibilia, M.; Wagner, E.F. Strain-dependent epithelial defects in mice lacking the EGF receptor. Science 1995, 269, 234–238. [Google Scholar] [CrossRef] [PubMed]
  163. Threadgill, D.W.; Dlugosz, A.A.; Hansen, L.A.; Tennenbaum, T.; Lichti, U.; Yee, D.; LaMantia, C.; Mourton, T.; Herrup, K.; Harris, R.C. Targeted disruption of mouse EGF receptor: Effect of genetic background on mutant phenotype. Science 1995, 269, 230–234. [Google Scholar] [CrossRef]
  164. Luetteke, N.C.; Qiu, T.H.; Fenton, S.E.; Troyer, K.L.; Riedel, R.F.; Chang, A.; Lee, D.C. Targeted inactivation of the EGF and amphiregulin genes reveals distinct roles for EGF receptor ligands in mouse mammary gland development. Development 1999, 126, 2739–2750. [Google Scholar] [CrossRef] [PubMed]
  165. Yan, Y.C.; Sun, Y.P.; Zhang, M.L.; Koide, S.S. Testis epidermal growth factor and spermatogenesis. Arch. Androl. 1998, 40, 133–146. [Google Scholar] [CrossRef]
  166. Manna, P.R.; Huhtaniemi, I.T.; Wang, X.J.; Eubank, D.W.; Stocco, D.M. Mechanisms of epidermal growth factor signaling: Regulation of steroid biosynthesis and the steroidogenic acute regulatory protein in mouse Leydig tumor cells. Biol. Reprod. 2002, 67, 1393–1404. [Google Scholar] [CrossRef] [PubMed]
  167. Ascoli, M.; Euffa, J.; Segaloff, D.L. Epidermal growth factor activates steroid biosynthesis in cultured Leydig tumor cells without affecting the levels of cAMP and potentiates the activation of steroid biosynthesis by choriogonadotropin and cAMP. J. Biol. Chem. 1987, 262, 9196–9203. [Google Scholar] [CrossRef]
  168. Evaul, K.; Hammes, S.R. Cross-talk between G protein-coupled and epidermal growth factor receptors regulates gonadotropin-mediated steroidogenesis in Leydig cells. J. Biol. Chem. 2008, 283, 27525–27533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Sordoillet, C.; Chauvin, M.A.; de Peretti, E.; Morera, A.M.; Benahmed, M. Epidermal growth factor directly stimulates steroidogenesis in primary cultures of porcine Leydig cells: Actions and sites of action. Endocrinology 1991, 128, 2160–2168. [Google Scholar] [CrossRef]
  170. Verhoeven, G.; Cailleau, J. Stimulatory effects of epidermal growth factor on steroidogenesis in Leydig cells. Mol. Cell. Endocrinol. 1986, 47, 99–106. [Google Scholar] [CrossRef]
  171. Syed, V.; Khan, S.A.; Nieschlag, E. Epidermal growth factor stimulates testosterone production of human Leydig cells in vitro. J. Endocrinol. Investig. 1991, 14, 93–97. [Google Scholar] [CrossRef]
  172. Hsueh, A.J.; Welsh, T.H.; Jones, P.B. Inhibition of ovarian and testicular steroidogenesis by epidermal growth factor. Endocrinology 1981, 108, 2002–2004. [Google Scholar] [CrossRef]
  173. Li, X.; Wang, Y.; Zhu, Q.; Yuan, K.; Su, Z.; Ge, F.; Ge, R.S.; Huang, Y. Epidermal growth factor regulates the development of stem and progenitor Leydig cells in rats. J. Cell. Mol. Med. 2020, 24, 7313–7330. [Google Scholar] [CrossRef] [PubMed]
  174. Tfelt-Hansen, J.; Yano, S.; John Macleod, R.; Smajilovic, S.; Chattopadhyay, N.; Brown, E.M. High calcium activates the EGF receptor potentially through the calcium-sensing receptor in Leydig cancer cells. Growth Factors 2005, 23, 117–123. [Google Scholar] [CrossRef] [PubMed]
  175. Guo, H.; Zhang, D.; Zhou, Y.; Sun, L.; Li, C.; Luo, X.; Liu, J.; Cui, S. Casein kinase 1α regulates testosterone synthesis and testis development in adult mice. Endocrinology 2023, 164, bqad042. [Google Scholar] [CrossRef] [PubMed]
  176. Jensen, M.B. Vitamin D and male reproduction. Nat. Rev. Endocrinol. 2014, 10, 175–186. [Google Scholar] [CrossRef]
  177. Morris, H.A.; Anderson, P.H. Autocrine and paracrine actions of vitamin D. Clin. Biochem. Rev. 2010, 31, 129–138. [Google Scholar]
  178. Adamczewska, D.; Słowikowska-Hilczer, J.; Walczak-Jędrzejowska, R. The association between vitamin D and the components of male fertility: A systematic review. Biomedicines 2023, 11, 90. [Google Scholar] [CrossRef]
  179. Haussler, M.R.; Jurutka, P.W.; Mizwicki, M.; Norman, A.W. Vitamin D receptor (VDR)-mediated actions of 1α,25(OH)2vitamin D3: Genomic and non-genomic mechanisms. Best Pract. Res. Clin. Endocrinol. Metab. 2011, 25, 543–559. [Google Scholar] [CrossRef]
  180. Hewison, M. Vitamin D and immune function: Autocrine, paracrine or endocrine? Scand. J. Clin. Lab. Investig. 2012, 72, 92–102. [Google Scholar] [CrossRef]
  181. Boisen, I.M.; Bøllehuus Hansen, L.; Mortensen, L.J.; Lanske, B.; Juul, A.; Blomberg Jensen, M. Possible influence of vitamin D on male reproduction. J. Steroid Biochem. Mol. Biol. 2017, 173, 215–222. [Google Scholar] [CrossRef]
  182. Foresta, C.; Strapazzon, G.; De Toni, L.; Perilli, L.; Di Mambro, A.; Muciaccia, B.; Sartori, L.; Selice, R. Bone mineral density and testicular failure: Evidence for a role of vitamin D 25-hydroxylase in human testis. J. Clin. Endocrinol. Metab. 2011, 96, 646–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  183. Foresta, C.; Selice, R.; De Toni, L.; Di Mambro, A.; Carraro, U.; Plebani, M.; Garolla, A. Altered bone status in unilateral testicular cancer survivors: Role of CYP2R1 and its luteinizing hormone-dependency. J. Endocrinol. Investig. 2013, 36, 379–384. [Google Scholar] [CrossRef]
  184. Anderson, P.H.; Hendrix, I.; Sawyer, R.K.; Zarrinkalam, R.; Manavis, J.; Sarvestani, G.T.; May, B.K.; Morris, H.A. Co-expression of CYP27B1 enzyme with the 1.5 kb CYP27B1 promoter-luciferase transgene in the mouse. Mol. Cell. Endocrinol. 2008, 285, 1–9. [Google Scholar] [CrossRef] [PubMed]
  185. Hendrix, I.; Anderson, P.; May, B.; Morris, H. Regulation of gene expression by the CYP27B1 promoter-study of a transgenic mouse model. J. Steroid Biochem. Mol. Biol. 2004, 89–90, 139–142. [Google Scholar] [CrossRef]
  186. Blomberg Jensen, M.; Nielsen, J.E.; Jørgensen, A.; Rajpert-De Meyts, E.; Kristensen, D.M.; Jørgensen, N.; Skakkebaek, N.E.; Juul, A.; Leffers, H. Vitamin D receptor and vitamin D metabolizing enzymes are expressed in the human male reproductive tract. Hum. Reprod. 2010, 25, 1303–1311. [Google Scholar] [CrossRef] [Green Version]
  187. Blomberg Jensen, M.; Jørgensen, A.; Nielsen, J.E.; Bjerrum, P.J.; Skalkam, M.; Petersen, J.H.; Egeberg, D.L.; Bangsbøll, S.; Andersen, A.N.; Skakkebæk, N.E.; et al. Expression of the vitamin D metabolizing enzyme CYP24A1 at the annulus of human spermatozoa may serve as a novel marker of semen quality. Int. J. Androl. 2012, 35, 499–510. [Google Scholar] [CrossRef]
  188. Holt, R.; Juel Mortensen, L.; Harpelunde Poulsen, K.; Nielsen, J.E.; Frederiksen, H.; Jørgensen, N.; Jørgensen, A.; Juul, A.; Blomberg Jensen, M. Vitamin D and sex steroid production in men with normal or impaired Leydig cell function. J. Steroid Biochem. Mol. Biol. 2020, 199, 105589. [Google Scholar] [CrossRef]
  189. Ferlin, A.; Selice, R.; Carraro, U.; Foresta, C. Testicular function and bone metabolism-Beyond testosterone. Nat. Rev. Endocrinol. 2013, 9, 548–554. [Google Scholar] [CrossRef]
  190. Lerchbaum, E.; Trummer, C.; Theiler-Schwetz, V.; Kollmann, M.; Wölfler, M.; Heijboer, A.C.; Pilz, S.; Obermayer-Pietsch, B. Effects of vitamin D supplementation on androgens in men with low testosterone levels: A randomized controlled trial. Eur. J. Nutr. 2019, 58, 3135–3146. [Google Scholar] [CrossRef] [Green Version]
  191. Tak, Y.J.; Lee, J.G.; Kim, Y.J.; Park, N.C.; Kim, S.S.; Lee, S.; Cho, B.M.; Kong, E.H.; Jung, D.W.; Yi, Y.H. Serum 25-hydroxyvitamin D levels and testosterone deficiency in middle-aged Korean men: A cross-sectional study. Asian J. Androl. 2015, 17, 324–328. [Google Scholar] [CrossRef]
  192. Canguven, O.; Talib, R.A.; El Ansari, W.; Yassin, D.-J.; Al Naimi, A. Vitamin D treatment improves levels of sexual hormones, metabolic parameters and erectile function in middle-aged vitamin D deficient men. Aging Male 2017, 20, 9–16. [Google Scholar] [CrossRef] [PubMed]
  193. Santos, H.O.; Howell, S.; Nichols, K.; Teixeira, F.J. Reviewing the evidence on vitamin D supplementation in the management of testosterone status and its effects on male reproductive system (testis and prostate): Mechanistically dazzling but clinically disappointing. Clin. Ther. 2020, 42, e101–e114. [Google Scholar] [CrossRef] [PubMed]
  194. Rafiq, R.; van Schoor, N.M.; Sohl, E.; Zillikens, M.C.; Oosterwerff, M.M.; Schaap, L.; Lips, P.; de Jongh, R.T. Associations of vitamin D status and vitamin D-related polymorphisms with sex hormones in older men. J. Steroid Biochem. Mol. Biol. 2016, 164, 11–17. [Google Scholar] [CrossRef]
  195. Panda, D.K.; Miao, D.; Tremblay, M.L.; Sirois, J.; Farookhi, R.; Hendy, G.N.; Goltzman, D. Targeted ablation of the 25-hydroxyvitamin D 1α-hydroxylase enzyme: Evidence for skeletal, reproductive, and immune dysfunction. Proc. Natl. Acad. Sci. USA 2001, 98, 7498–7503. [Google Scholar] [CrossRef]
  196. Blomberg Jensen, M.; Lieben, L.; Nielsen, J.E.; Willems, A.; Jørgensen, A.; Juul, A.; Toppari, J.; Carmeliet, G.; Rajpert-De Meyts, E. Characterization of the testicular, epididymal and endocrine phenotypes in the Leuven Vdr-deficient mouse model: Targeting estrogen signalling. Mol. Cell. Endocrinol. 2013, 377, 93–102. [Google Scholar] [CrossRef] [PubMed]
  197. Wang, L.; Lu, H.; Wang, S.; Liu, H.; Guo, M.; Bai, H.; Zeng, W.; Zhang, T. Vitamin D receptor affects male mouse fertility via regulation of lipid metabolism and testosterone biosynthesis in testis. Gene 2022, 834, 146589. [Google Scholar] [CrossRef]
  198. Zhang, T.; Yin, Y.; Liu, H.; Du, W.; Ren, C.; Wang, L.; Lu, H.; Zhang, Z. Generation of VDR knock-out mice via zygote injection of CRISPR/Cas9 system. PLoS ONE 2016, 11, e0163551. [Google Scholar] [CrossRef] [Green Version]
  199. Johnson, L.E.; DeLuca, H.F. Vitamin D receptor null mutant mice fed high levels of calcium are fertile. J. Nutr. 2001, 131, 1787–1791. [Google Scholar] [CrossRef] [Green Version]
  200. Kinuta, K.; Tanaka, H.; Moriwake, T.; Aya, K.; Kato, S.; Seino, Y. Vitamin D is an important factor in estrogen biosynthesis of both female and male gonads. Endocrinology 2000, 141, 1317–1324. [Google Scholar] [CrossRef]
  201. Xue, Z.; Zhuang, J.; Bai, H.; Wang, L.; Lu, H.; Wang, S.; Zeng, W.; Zhang, T. VDR mediated HSD3B1 to regulate lipid metabolism and promoted testosterone synthesis in mouse Leydig cells. Genes Genom. 2022, 44, 583–592. [Google Scholar] [CrossRef]
  202. Zamani, A.; Saki, F.; Hatami, N.; Koohpeyma, F. Stereological assessment of the effects of vitamin D deficiency on the rat testis. BMC Endocr. Disord. 2020, 20, 162. [Google Scholar] [CrossRef] [PubMed]
  203. Hauschka, P.V.; Lian, J.B.; Cole, D.E.; Gundberg, C.M. Osteocalcin and matrix Gla protein: Vitamin K-dependent proteins in bone. Physiol. Rev. 1989, 69, 990–1047. [Google Scholar] [CrossRef] [PubMed]
  204. Poser, J.W.; Esch, F.S.; Ling, N.C.; Price, P.A. Isolation and sequence of the vitamin K-dependent protein from human bone. Undercarboxylation of the first glutamic acid residue. J. Biol. Chem. 1980, 255, 8685–8691. [Google Scholar] [CrossRef] [PubMed]
  205. Oury, F.; Ferron, M.; Huizhen, W.; Confavreux, C.; Xu, L.; Lacombe, J.; Srinivas, P.; Chamouni, A.; Lugani, F.; Lejeune, H.; et al. Osteocalcin regulates murine and human fertility through a pancreas-bone-testis axis. J. Clin. Investig. 2013, 123, 2421–2433. [Google Scholar] [CrossRef] [PubMed]
  206. Khrimian, L.; Obri, A.; Ramos-Brossier, M.; Rousseaud, A.; Moriceau, S.; Nicot, A.-S.; Mera, P.; Kosmidis, S.; Karnavas, T.; Saudou, F.; et al. Gpr158 mediates osteocalcin’s regulation of cognition. J. Exp. Med. 2017, 214, 2859–2873. [Google Scholar] [CrossRef]
  207. Oury, F.; Sumara, G.; Sumara, O.; Ferron, M.; Chang, H.; Smith, C.E.; Hermo, L.; Suarez, S.; Roth, B.L.; Ducy, P.; et al. Endocrine regulation of male fertility by the skeleton. Cell 2011, 144, 796–809. [Google Scholar] [CrossRef] [Green Version]
  208. Pi, M.; Chen, L.; Huang, M.Z.; Zhu, W.; Ringhofer, B.; Luo, J.; Christenson, L.; Li, B.; Zhang, J.; Jackson, P.D.; et al. GPRC6A null mice exhibit osteopenia, feminization and metabolic syndrome. PLoS ONE 2008, 3, e3858. [Google Scholar] [CrossRef]
  209. Shan, C.; Yue, J.; Liu, W. Broadening the role of osteocalcin in the hypothalamic-pituitary-gonadal axis. J. Endocrinol. 2021, 249, R43–R51. [Google Scholar] [CrossRef]
  210. Hannemann, A.; Breer, S.; Wallaschofski, H.; Nauck, M.; Baumeister, S.E.; Barvencik, F.; Amling, M.; Schinke, T.; Haring, R.; Keller, J. Osteocalcin is associated with testosterone in the general population and selected patients with bone disorders. Andrology 2013, 1, 469–474. [Google Scholar] [CrossRef] [Green Version]
  211. Jawich, K.; Rocca, M.S.; Al Fahoum, S.; Alhalabi, M.; Di Nisio, A.; Foresta, C.; Ferlin, A.; De Toni, L. RS 2247911 polymorphism of GPRC6A gene and serum undercarboxylated-osteocalcin are associated with testis function. J. Endocrinol. Investig. 2022, 45, 1673–1682. [Google Scholar] [CrossRef]
  212. De Toni, L.; De Filippis, V.; Tescari, S.; Ferigo, M.; Ferlin, A.; Scattolini, V.; Avogaro, A.; Vettor, R.; Foresta, C. Uncarboxylated osteocalcin stimulates 25-hydroxy vitamin D production in Leydig cell line through a GPRC6a-dependent pathway. Endocrinology 2014, 155, 4266–4274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Key pathways in LH-mediated steroidogenesis in Leydig cells. Upon LH binding to LHCGR, multiple pathways are activated: (i) LHCGR activation stimulates adenylate cyclase (AC), increasing the conversion of ATP to cyclic AMP (cAMP). Elevated cAMP levels activate protein kinase A (PKA) by releasing its catalytic subunits (C) from its regulatory subunits (R). (ii) LHCGR activation triggers phospholipase C (PLC), resulting in the breakdown of phosphatidylinositol 4,5-bisphosphate (PIP2) in diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3). DAG activates protein kinase C (PKC), while IP3 binds to its receptor (IP3R) in the endoplasmic reticulum (ER), leading to calcium (Ca2+) release. The ryanodine receptor (RyR) in the ER also facilitates Ca2+ release. Calcium can also enter the cell through plasma membrane channels. Calcium-bound calmodulin (CaM) activates calcium/calmodulin-dependent protein kinase kinase I (CAMKKI), which phosphorylates calcium/calmodulin-dependent protein kinase I (CAMKI). (iii) LH binding to its receptor activates Ras, subsequently activating the MAPKKK Raf. This activation triggers the phosphorylation of MAPKKs (MEK1/2) and MAPKs (ERK1/2). (iv) The activation of LHCGR activates phosphatidylinositol-3-kinase (PI3K), which phosphorylates protein kinase B (PKB). Once activated, these kinases translocate to the nucleus (indicated by grey dashed arrows), where they phosphorylate various transcription factors, such as COUP-TFI, COUP-TFII, GATA4, CREB, STAT5A, STAT5B, NOR1, NURR1, JUN and NUR77. This leads to the upregulated expression of genes involved in steroidogenesis and, ultimately, increased testosterone production. See text for references.
Figure 1. Key pathways in LH-mediated steroidogenesis in Leydig cells. Upon LH binding to LHCGR, multiple pathways are activated: (i) LHCGR activation stimulates adenylate cyclase (AC), increasing the conversion of ATP to cyclic AMP (cAMP). Elevated cAMP levels activate protein kinase A (PKA) by releasing its catalytic subunits (C) from its regulatory subunits (R). (ii) LHCGR activation triggers phospholipase C (PLC), resulting in the breakdown of phosphatidylinositol 4,5-bisphosphate (PIP2) in diacylglycerol (DAG) and inositol 1,4,5-triphosphate (IP3). DAG activates protein kinase C (PKC), while IP3 binds to its receptor (IP3R) in the endoplasmic reticulum (ER), leading to calcium (Ca2+) release. The ryanodine receptor (RyR) in the ER also facilitates Ca2+ release. Calcium can also enter the cell through plasma membrane channels. Calcium-bound calmodulin (CaM) activates calcium/calmodulin-dependent protein kinase kinase I (CAMKKI), which phosphorylates calcium/calmodulin-dependent protein kinase I (CAMKI). (iii) LH binding to its receptor activates Ras, subsequently activating the MAPKKK Raf. This activation triggers the phosphorylation of MAPKKs (MEK1/2) and MAPKs (ERK1/2). (iv) The activation of LHCGR activates phosphatidylinositol-3-kinase (PI3K), which phosphorylates protein kinase B (PKB). Once activated, these kinases translocate to the nucleus (indicated by grey dashed arrows), where they phosphorylate various transcription factors, such as COUP-TFI, COUP-TFII, GATA4, CREB, STAT5A, STAT5B, NOR1, NURR1, JUN and NUR77. This leads to the upregulated expression of genes involved in steroidogenesis and, ultimately, increased testosterone production. See text for references.
Endocrines 04 00041 g001
Figure 2. Additional signaling pathways implicated in the stimulation of Leydig cell steroidogenesis. Several hormones and molecules contribute to increased steroid production in Leydig cells. Upon growth hormone (GH) binding to the GH-receptor (GHR), the receptor becomes activated, leading to the formation of GHR dimers. Prolactin receptor (PRLR) is also activated by prolactin (PRL) binding. Both active GHR and PRLR induce the activation of JAKs, which phosphorylate STAT5A and STAT5B transcription factors. Phosphorylated STAT5s hetero- and homodimerize, and translocate to the nucleus where they regulate gene transcription. In addition to GH, other growth factors involved in steroidogenesis are insulin (INS), insulin-like growth factor 1 (IGF1), fibroblast growth factor (FGF), and epidermal growth factor (EGF). Insulin and IGF1 actions are mediated through the activation of two related receptors: INS receptor (INSR) and IGF1 receptor (IGF1R). Both INS and IGF1 can bind to both receptors, albeit with different affinities. INS, IGF1, FGF9, and EGF trigger a series of events upon binding to its tyrosine kinase receptors, resulting in the activation of at least two pathways, phosphatidylinositol-3-kinase (PI3K)/protein kinase B (PKB) and MAPKs. Conversely, high levels of insulin activate DAX1/NR0B1, indicated by red dashed arrows, which represses hormone-induced steroidogenesis. FGF9 can also activate the protein kinase A (PKA) pathway. EGF receptor (EGFR) can activate protein kinase C (PKC), cJun N-terminal kinases (JNK) and casein kinase 1α (CK1α). In addition, undercarboxylated osteocalcin (unOCN) binds to and activates G protein-coupled receptor family C group 6-member A (GPRC6A) coupled to adenylate cyclase (AC). This leads to increased cAMP production, activation of PKA, and subsequent activation of the downstream transcription factor, CREB. Activated vitamin D (1,25(OH)2D) acts through two pathways: non-genomic and genomic. In the non-genomic pathway, 1,25(OH)2D binds to the vitamin D receptor (VDR) on the cell membrane, activating phospholipases, second messengers, and kinases. In the genomic pathway, 1,25(OH)2D enters the cell, binds to the VDR in the cytoplasm, and forms a complex with the retinoid X receptor (RXR). This complex acts as a transcription factor, recognizing vitamin D response elements (VDREs) in target gene promoter regions. See text for references.
Figure 2. Additional signaling pathways implicated in the stimulation of Leydig cell steroidogenesis. Several hormones and molecules contribute to increased steroid production in Leydig cells. Upon growth hormone (GH) binding to the GH-receptor (GHR), the receptor becomes activated, leading to the formation of GHR dimers. Prolactin receptor (PRLR) is also activated by prolactin (PRL) binding. Both active GHR and PRLR induce the activation of JAKs, which phosphorylate STAT5A and STAT5B transcription factors. Phosphorylated STAT5s hetero- and homodimerize, and translocate to the nucleus where they regulate gene transcription. In addition to GH, other growth factors involved in steroidogenesis are insulin (INS), insulin-like growth factor 1 (IGF1), fibroblast growth factor (FGF), and epidermal growth factor (EGF). Insulin and IGF1 actions are mediated through the activation of two related receptors: INS receptor (INSR) and IGF1 receptor (IGF1R). Both INS and IGF1 can bind to both receptors, albeit with different affinities. INS, IGF1, FGF9, and EGF trigger a series of events upon binding to its tyrosine kinase receptors, resulting in the activation of at least two pathways, phosphatidylinositol-3-kinase (PI3K)/protein kinase B (PKB) and MAPKs. Conversely, high levels of insulin activate DAX1/NR0B1, indicated by red dashed arrows, which represses hormone-induced steroidogenesis. FGF9 can also activate the protein kinase A (PKA) pathway. EGF receptor (EGFR) can activate protein kinase C (PKC), cJun N-terminal kinases (JNK) and casein kinase 1α (CK1α). In addition, undercarboxylated osteocalcin (unOCN) binds to and activates G protein-coupled receptor family C group 6-member A (GPRC6A) coupled to adenylate cyclase (AC). This leads to increased cAMP production, activation of PKA, and subsequent activation of the downstream transcription factor, CREB. Activated vitamin D (1,25(OH)2D) acts through two pathways: non-genomic and genomic. In the non-genomic pathway, 1,25(OH)2D binds to the vitamin D receptor (VDR) on the cell membrane, activating phospholipases, second messengers, and kinases. In the genomic pathway, 1,25(OH)2D enters the cell, binds to the VDR in the cytoplasm, and forms a complex with the retinoid X receptor (RXR). This complex acts as a transcription factor, recognizing vitamin D response elements (VDREs) in target gene promoter regions. See text for references.
Endocrines 04 00041 g002
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

de Mattos, K.; Pierre, K.J.; Tremblay, J.J. Hormones and Signaling Pathways Involved in the Stimulation of Leydig Cell Steroidogenesis. Endocrines 2023, 4, 573-594. https://doi.org/10.3390/endocrines4030041

AMA Style

de Mattos K, Pierre KJ, Tremblay JJ. Hormones and Signaling Pathways Involved in the Stimulation of Leydig Cell Steroidogenesis. Endocrines. 2023; 4(3):573-594. https://doi.org/10.3390/endocrines4030041

Chicago/Turabian Style

de Mattos, Karine, Kenley Joule Pierre, and Jacques J. Tremblay. 2023. "Hormones and Signaling Pathways Involved in the Stimulation of Leydig Cell Steroidogenesis" Endocrines 4, no. 3: 573-594. https://doi.org/10.3390/endocrines4030041

Article Metrics

Back to TopTop