Skip to main content

Pharmacogenomic of LH and its receptor: are we ready for clinical practice?

Abstract

Luteinizing hormone (LH) is fundamental to support development and reproduction. It acts through a receptor expressed in the gonads, modulating mitogenic, anti-apoptotic, and steroidogenic signals. LH is also marketed as a drug for controlled ovarian stimulation (COS), where it is administered to women to support the action of follicle-stimulating hormone and can lead to specific responses, depending on the individual genetic background. These concepts underline the relevance of a pharmacogenetic approach to COS, in the attempt to optimize clinical outcomes and avoid adverse events. However, knowledge is currently limited by the paucity of clinical studies. This review aims to provide a comprehensive overview of LH and its receptor activity, starting from the description of their molecular pathways from in vitro studies. Data on LH action from in vivo studies were described, as well as the impact of LH and LH/choriogonadotropin (hCG) receptor genetic variants on folliculogenesis and its association with infertility or polycystic ovarian syndrome. Finally, evidence from clinical studies evaluating genetic polymorphisms in the context of assisted reproductive technology treatments and its implications for a pharmacogenomic approach were discussed.

Introduction

Luteinizing hormone (LH) is a gonadotropin fundamental to development and reproduction. It is a dimeric glycoprotein released in a pulsatile fashion by the pituitary gland and acts through the LH/choriogonadotropin (hCG) receptor (LHCGR), which is expressed in the gonads [1]. LH has a β subunit (LHβ), specific for receptor binding, and an α subunit shared with other structurally similar glycoproteins: follicle-stimulating hormone (FSH), thyroid-stimulating hormone (TSH), and hCG [2]. The β-subunit is coded by the LHB gene located in the genomic locus 19q13.33, while the α-subunit is coded by the CGA gene, located at 6q14.3, and is assembled with LHβ within pituitary gonadotropic cells. The final product is a ~ 33 kDa molecule that, in women of fertile age, exerts a key role in supporting the production of ovarian sex steroids and modulating mitogenic and anti-apoptotic signals. These functions sustain follicular growth, oocyte maturation, and ovulation, as well as the luteinization of granulosa cells.

LHCGR is coded by the homonym gene, located in the genomic position 2p16.3, which spans about 80 kbases with 11 exons and 10 introns, and consists in a common receptor for both LH and hCG [3]. These two ligands have different physiological functions: hCG is the pregnancy hormone, produced by trophoblast cells to induce progesterone production; hCGβ molecules are coded by cluster genes (CGBs), located in proximity of LHB and likely evolved from a common ancestral sequence [4]. However, hCGβ has an additional carboxyl-terminal peptide of about 30 amino acids carrying six glycosylation sites, and an extended half-life [5].

Although LH and hCG display these physiological, structural, and biochemical differences, they are marketed as recombinant or extractive and highly purified drugs to support FSH in controlled ovarian stimulation (COS), in the context of assisted reproduction [6]. The action of these molecules is modulated by hormone and receptor polymorphic variants that influence individual response to COS, as well as predisposition to diseases and adverse events [7]. The pharmacogenomic approach could represent an opportunity to improve the efficiency of COS [8]. Apart from FSH, it is widely acknowledged that LH is crucial for folliculogenesis [9, 10]. Several studies have demonstrated that a specific subset of women undergoing IVF treatment – namely, those with low prognosis according to POSEIDON criteria – may benefit from LH during COS [11,12,13]. Moreover, there are specific genetic variants of LH and its receptor that could benefit from FSH or LH dose adjustment during assisted reproductive therapy (ART) [14,15,16]. These concepts form the basis of the pharmacogenetic approach to assisted reproduction; this review provides a summary of the molecular pathways and pharmacogenetics of LH in clinical practice. Moreover, results from in vivo and in vitro studies about LH signaling, LHβ and LHCGR variants, as well as their clinical impact, were discussed.

Materials and methods

We performed a literature search in PubMed, Scopus, Embase, and the ISI Web of Science database. The search terms were: ‘LHCGR’, ‘LH’, ‘polymorphisms’, ‘genetic variants’, ‘ART’, ‘IVF’, ‘polycystic ovarian syndrome’, and ‘PCOS’ from the inception to January 2024. The most relevant studies that analyzed the impact of LH or LHCGR polymorphism on IVF outcomes are summarized in Supplemental Table 1. No language or time restriction was adopted.

Results

Molecular pathways involved in LH signaling results from in vitro studies

LHCGR is a 7-transmembrane, class A, G protein-coupled receptor (GPCR) [1]; its active and inactive conformations have been recently resolved by cryogenic electron microscopy [17]. Hormone binding to the receptor extracellular domain induces a ‘push/pull’ movement involving the hinge region and impacting the spatial arrangement of the transmembrane stretches [17], resulting in the activation of multiple intracellular signaling cascades [3]. It is generally accepted that LHCGR mediates both cAMP and intracellular Ca2+ increase, as well as sex steroid hormone production. These molecules were detectable using the first, pioneering assays available, such as radioimmunoassay, and have long been considered the main players involved in LH/hCG signaling [18, 19].

cAMP and Ca2+ increase rapidly and belong to two separate signaling pathways mediated by different G proteins: Gαs and Gαq, respectively [20]. cAMP is a second messenger inducing the activation of PKA and the phosphorylation of CREB, before being metabolized to AMP [21]. In granulosa cells, relatively high intracellular cAMP concentrations have been linked to pro-apoptotic effects [22,23,24] and, at the same time, to the compartmentalization of progesterone synthesis and androgen conversion to estrogens [25].

Androgens, mainly androstenedione, are produced by theca cells upon binding of phosphorylated CREB (pCREB) to CRE DNA target sequences. This is a PKA-dependent event that induces the transcription of steroidogenic enzyme-coding genes such as steroidogenic acute regulatory protein (STARD1), cytochrome P450 family 17 subfamily A member 1 (CYP17A1), and aromatase (CYP19A1) [26]. PKA activation is also followed by phosphorylation of the extracellular-regulated kinase 1/2 (ERK1/2), accompanying the inhibition of progesterone production and stimulation of androgens synthesis [27], and upregulating mitogenic processes in gonadal steroidogenic cells [28, 29]. Moreover, ERK1/2 phosphorylation is linked to the downregulation of receptor mRNA transcripts [28] and activation of GPCR kinases responsible for receptor phosphorylation and internalization into intracellular vesicles [29]. In particular, the compartmentalization of LHCGR is mediated by β-arrestins, which are proteins responsible for a second wave of ERK1/2 phosphorylation [30], occurring possibly as an opposing effect to cAMP-dependent pro-apoptotic signals [31] (Fig. 1).

Fig. 1
figure 1

Main LHCGR-mediated signaling pathways. Upon hormone binding, LHCGR activates multiple signaling pathways via G protein and B-arrestin recruitment. These signaling cascades converge mainly into steroidogenic and pro-apoptotic stimuli, counterbalanced by mitogenic and anti-apoptotic signals

In vitro experiments in transfected cells, expressing the murine receptor, demonstrated that gonadotropins are responsible for Gαq protein and phospholipase C (PLC) activation, inositol trisphosphate (IP3) binding to calcium channels of endoplasmic reticulum, and mobilization of intracellular Ca2+ [20]. Calcium ions modulate the activity of protein calmodulin kinases, a key event to control cell proliferation [32] and transport of cholesterol into the mitochondrion, as a rate-limiting step for steroidogenesis [33, 34]. Interestingly, the half-maximal (EC50) hCG concentration activating the cAMP/PKA pathway is ~ 20 times lower than that required to trigger PLC/Ca2+ pathway activation, suggesting that LHCGR has dual signaling potential [35].

Recent scientific advancements have demonstrated the existence of multiple intracellular signaling cascades, providing a more detailed picture of LH- and hCG-mediated signals. For instance, the βγ dimer of G protein may indirectly lead to protein kinase B (AKT) activation, which upregulates survival signals [36], inhibits aromatase [37] and supports STARD1 expression [38]. The preferential activation of specific signaling patterns depends on several factors. First, LHCGR signaling may be allosterically modulated by other 7-transmembrane partners, which can physically interact with the receptor to form heteromeric assemblies in the cell surface [22, 39,40,41]. Second, prevalent β-arrestin/ERK1/2 and AKT activation was found at low receptor density [42, 43], while marked cAMP activation would be due to increased Gαs coupling occurring at relatively high receptor expression levels [22, 23]. These aspects shed light on the physiological impact of extremely variable gonadotropin receptor levels throughout the human menstrual cycle [44]. Agonist binding induces receptor aggregation [45] and sequestration from the cell surface [46], preceding its internalization in endosomal vesicles [47]. In particular, LHCGR internalization is mediated by GPCR kinases and other modulators, such as β-arrestins, that may form super-complexes of signaling modules sustaining prolonged cAMP [48, 49] and direct ERK1/2 activation [23].

Taken together, LHCGR mediates a complex, spatial-temporal network of multiple signaling pathways triggered by LH and hCG. The two hormones act as different ligands linked to specific signaling patterns [3], increasing the complexity of the picture. LH binding to LHCGR leads to preferential activation of proliferative and anti-apoptotic signals delivered through ERK1/2 and AKT, essential to ovarian follicle growth and maturation, while hCG is a potent progestational based on its physiological role [50,51,52,53,54]. These data illustrate the different activity exerted by LH and hCG in vitro, which was confirmed also in a clinical setting [55], and suggest the existence of natural, specific regulatory mechanisms adapting gonadotropin signals to physiological requirements (steroidogenesis, cell death, or survival).

Knowledge about LH action from in vivo studies

Recent decades have seen the generation of mutant hormone/receptor mice, allowing LH functions to be elucidated in vivo. Results from these studies must be interpreted with caution, since animal models for gonadotropin functioning, especially multi-ovulatory species, cannot be fully representative of human physiology.

One of the first genetically modified mice overexpressed CGB and CGA [56]. These animals were developed expecting to resemble the phenotype of humans with activating LHCGR mutations (i.e., males affected by testotoxicosis, tumorigenesis, or precocious puberty) and overall asymptomatic females. Instead, transgenic male mice had normal phenotypes, whereas females displayed luteinized ovarian follicles with hemorrhagic cysts and luteomas, precocious puberty, obesity, and other non-reproductive features, such as pituitary adenomas, mammary tumors, and pseudopregnancy [56, 57]. An overall similar phenotype was obtained in female mice overexpressing a chimeric LHβ fused with the carboxyl-terminal peptide of hCG. This molecule has additional glycosylations, extending its half-life, and can lead to polycystic ovaries, relatively high levels of sex steroids, infertility, and ovarian tumors [58].

Knockout mice were developed, starting with the deletion of the CGA gene. These animals had obvious hypogonadism and hypothyroidism [59], due to the lack of full LH molecules, as well as FSH and TSH, which share the same α subunit. LHB knockout mice were also developed, reflecting this condition in humans. Male mice were affected by hypogonadism and displayed very low androgen levels and hypoplastic Leydig cells despite overall normal FSH levels, while females showed anovulation and collapse of antral follicles, and were infertile [60].

LH receptor knockout mouse models (LuRKO) have phenotypes similar to the LHβ knockout, although they are not fully representative of the condition in humans, where full inactivation of LHCGR leads to type-1 Leydig cell hypoplasia and hermaphroditism. In mice at the fetal stage, the production of testosterone can occur through a gonadotropin-independent pathway [61], stimulating fetal Leydig cells even in the absence of LH or its receptor, given the support of other paracrine factors [62]. Therefore, LuRKO mice display impaired sexual maturation but have similar phenotypes of the wild-type at birth. Interestingly, the effects of the absence of LH receptors may be partially rescued in older mice (≥ 12 months), where LH-independent testosterone production could support qualitatively, but not quantitatively, similar spermatogenesis to wild-type mice [63]. In LuRKO mice, similar effects were obtained by FSH receptor (FSHR) activating mutations, where spermatogenesis is rescued even in the presence of the anti-androgen flutamide [64].

These findings suggest the existence of Sertoli cell–dependent paracrine factors capable of supporting partially Leydig cell functions, reflective of partially overlapping spermatogenic pathways. Moreover, these data suggest that the hormonal regulation of spermatogenesis has shifted from the dominance of gonadotropins to sex steroids during evolution. However, these data were not fully replicated in female LuRKO mice, where the expression of constitutively active FSHR led to the progression of antral follicles to the preovulatory stage and enhanced estrogenic activity but failed to rescue the healthy phenotype from hypogonadism and anovulation [65]. Taken together, studies from in vivo models have confirmed the relevance of the LH/LHCGR system for reproduction, although potential translation of results to humans must take careful account of sex- and species-specific effects.

The impact of LH and LHCGR genetic variants on signal transduction during folliculogenesis

Few single-nucleotide polymorphisms (SNPs) within the LHB/CGB gene cluster have been described. In general, though little is known about CGB SNPs and their possible association with miscarriage [66], LHB SNPs are linked to human phenotypic variations that might mildly contribute to the pathogenesis of reproductive diseases, such as polycystic ovary syndrome (PCOS) [67]. The main molecular mechanism by which LHB and LHCGR SNPs impact ovarian functions would rely on the modulation of androgen production and, in turn, on the subsequent perturbation of the hypothalamic-pituitary-gonadal axis. However, it is plausible that mitogenic signals mediated directly by gonadotropins, fundamental to support gametogenesis, could also be perturbed. The endocrine adaptation to androgen levels is controlled by feedback mechanisms that lead to changes of serum LH and FSH, as well as levels of gonadal and adrenal steroids. Together, these events impact on gametogenesis and metabolism, as functions that, in large part, are directly or indirectly dependent on gonadotropins and steroids. However, clinical data are limited, and functional characterizations are mostly missing, suggesting that these SNPs do not substantially impact fertility [67].

One of the best-characterized LHB polymorphic variants was found in the Finnish population: V-LH, which consists in the double amino acid tryptophan–arginine and isoleucine–threonine change at positions 28 and 35 of the protein chain [68, 69]. It has an additional glycosylation site [70], lower half-life, and receptor binding, as well as decreased potency in activating progesterone and cAMP, than the classical LH form [71, 72]. The V-LH variant was associated with infertility in homozygous Japanese women [73], as well as with the worst outcome of intracytoplasmic sperm injection procedures, reflecting enhanced pro-apoptotic signals detected in vitro, such as caspase 3 cleavage and DNA fragmentation index [74]. However, these data were never extensively replicated by independent studies in different ethnic groups, suggesting that the impact of V-LH on the phenotype is overall weak. This hormone variant is less frequent in obese PCOS women than in non-obese PCOS and healthy women, indicating that it might be protective against certain metabolic features related to the disease [75], although a further investigation failed to confirm this association [76]. Interestingly, the relatively low hormone bioactivity is compensated by higher V-LH expression than LH, due to SNPs falling within the promoter region in linkage disequilibrium with those at positions 25 and 35, increasing its activity in vivo [77, 78]. Together, these data suggest that the V-LH consists of a polymorphic variant associated with overall mild phenotypes.

In women, some other LHB SNPs were associated with infertility [79] or central precocious puberty [80]. In particular, a SNP characterized by the synonymous amino acid T–C change within the exon 3 at gene sequence position 294, was found to be more frequent in South Indian women with PCOS compared with healthy controls [81]. Although the role of this SNP in PCOS pathogenesis is unknown, it was hypothesized that it could impact the function of a LHB palindromic gene RUVBL2, coding a protein involved in the regulation of DNA transcription [82]. Another LHB SNP of potential clinical interest is provided by the asparagine–serine change at position 312 (p.Asn312Ser) of the protein chain, which is close to a glycosylation site and could impact sensitivity to the hormone and live birth rate [15, 83, 84]. Although LHB polymorphic variants could be promising targets for future pharmacogenomic research, these results require confirmation by independent clinical studies in other populations and functional in vitro support.

Most SNPs modulating LH/hCG signaling are carried by LHCGR [3]. The receptor is a hot-spot for certain reproductive diseases, such as PCOS [85,86,87], reflecting the relevance of a fine-tuned regulation of LH signaling to support folliculogenesis. Although the mechanism of PCOS pathogenesis is still largely unclear, marked LHCGR-dependent signals are likely important. They could lead to excessive androgen production which, in turn, impacts the endocrine control of the hypothalamus-pituitary-gonadal axis, interfering with ovarian follicular maturation and metabolism [88]. In fact, several LHCGR SNPs were associated with PCOS in different populations [89,90,91,92,93,94,95,96]. For instance, the exon 10 SNP characterized by the alanine–serine change at position 312 was associated with serum LH levels in PCOS patients [97]. Interestingly, the same SNP was linked to spermatogenic damage and infertility in males [98], suggesting that the variant falls within a key region for receptor functioning. Beyond PCOS, the possible link between LHCGR SNPs and clinical outcome of assisted reproduction, such as oocyte/embryo quality, was also discussed [99]. Although these findings are encouraging, the LHCGR-dependent molecular mechanism at the basis of PCOS pathogenesis remains poorly understood and the role of the receptor as a potential target for pharmacological approach to the disease is under-researched [99]. The clinical effect of LHβ and LHCGR genetic variants, as well as of possible pharmacogenomic approaches, will be discussed in the next sections.

The clinical effect of LHB genetic variants

In COS, it has been suggested that elevated requirement of FSH may result from weak LHCGR activity. Therefore, these patients might benefit from exogenous LH administration rather than increased FSH dose. This effect could be linked to V-LH which, in the context of COS, cannot achieve adequate levels to compensate for its reduced bioactivity. In a retrospective analysis [100], where patients were divided into three groups according to the FSH dose required, the frequency of V-LH was higher in women with ovarian resistance to FSH administration in association with a lower number of oocytes retrieved. The LH versus V-LH genotypes were stratified in another multicentric study, demonstrating that elevated cumulative doses of FSH were associated with the V-LH genetic variant [16]. The mean number of oocytes retrieved, fertilization rate, and pregnancy rate did not differ between the two groups, indicating that the highest doses of FSH may counterbalance the negative impact of low V-LH bioactivity in inducing oocyte competence and impacting on IVF outcome. However, a significant reduction in the mean of embryo number transferred has been reported. The investigators posited an interplay between FSH- and LH-mediated signals to determine successful oocyte maturation and meiosis [16]. Recently, V-LH was associated with a lower pregnancy rate in gonadotropin-releasing hormone (GnRH) antagonists, but not in long GnRH agonist cycles; these differences were attributed to variations in clinical protocols [101]. Endogenous LH levels were lower with the antagonist cycle, compared with the agonist cycle [102,103,104]. Therefore, V-LH carriers are associated with reduced pregnancy rates only when they undergo profound LH suppression induced by antagonist protocols.

Another clinically interesting LHβ variant consists in the single missense exon 3 variation, consisting in the amino acid serine–glycine changes at position 102 (Gly1502Ser, rs1056917). This variant may change LH bioactivity, since a single study reported the association between the SNP and reduced LH level, and history of infertility [105]. However, another study prospectively enrolling 220 women undergoing long protocol for COS and IVF found no significant association between this SNP and ovarian response [106], suggesting that overall effects of this genetic variation are likely mild.

The clinical effect of LHCGR genetic variants

LHCGR exon 10 polymorphisms

One of the most studied polymorphisms is the exon 10 p.Asn312Ser. It is relatively common: in European Caucasians, the allele frequency is 41% asparagine (Asn) and 49% serine (Ser), compared with 68% Asn and 32% Ser in the Sub-Saharan African population (https://www.ncbi.nlm.nih.gov/snp/rs2293275). It was demonstrated that Asn-homozygous women required a lower amount of gonadotropin per day, during COS, than Ser-homozygous [84], while the latter have a fourfold higher chance of pregnancy than Asn-homozygous. In the same population, authors later reported significantly higher live birth and cumulative live birth rates in Ser-variant versus Asn-variant carriers [83]. This was explained by the increased number of good-quality embryos found in the Ser carrier group [83], according to the guidelines by Gardner and Schoolcraft [107, 108]. Consistently with Lindgren findings, in a prospective study involving 210 women, higher clinical pregnancy rate was observed in Ser-homozygous carriers than heterozygous women after fresh embryo transfer [109]. However, we obtained different results in a multicenter retrospective study involving 94 normogonadotropic women from three European IVF centers, where no significant association was found in terms of ovarian response (number of oocytes retrieved, MII oocytes) and pregnancy rate among different LHCGR haplotypes [110]. Our findings were corroborated by a recent analysis involving 1183 women, ages 18–40 years and undergoing their first assisted reproductive technology cycle, where the association between the LHCGR Asn312Ser variant and pregnancy rate was not detected [111]. Discrepancies between those studies could be linked to differences in their study designs, IVF protocols adopted, and ethnicity of participants.

Another exon 10 LHCGR polymorphism consists of the Asn–Ser change at position 291 (rs12470652). This variant was associated with increased receptor sensitivity [112] and has a prevalence of 5% in Europe (https://www.ncbi.nlm.nih.gov/snp/rs12470652). The clinical relevance of this polymorphism was assessed; no association was found with PCOS risk [113, 114], nor response to testicular cancer treatment [115]. In our multicenter prospective studies, we observed that this variant was associated with greater response to COS, evaluated as oocytes, and mature oocytes retrieved [110, 116]. In combination with other FSHR polymorphisms, this variant is associated with the ratio between cumulative FSH dose and total number of oocytes retrieved (odds ratio [OR] 5.44, 95% CI 3.18–7.71; p < 0.001), supporting the concept of varying sensitivity to the ligand depending on LHCGR Asn291Ser phenotype [110, 116]. These results were confirmed by recent research demonstrating that the Ser variant was significantly associated with PCOS risk, which is typically characterized by an increased sensitivity to exogenous gonadotropin during COS. Unfortunately, the evidence reported so far is limited due to the overall low prevalence of the Ser variant and a paucity of data on homozygotic carriers [113, 114, 116].

LHCGR A–G intronic nucleotide variation

Another LHCGR polymorphism consists in the A–G intronic nucleotide variation (rs13405728), which was strongly associated with PCOS and first described in a genome-wide association study in Chinese women [85]. The G allele occurs in 8% of the global population, with the highest prevalence in Africa (27–31%); https://www.ncbi.nlm.nih.gov/snp/rs13405728#frequency_tab). A meta-analysis revealed that the OR for developing PCOS was significantly lower in G than in A carriers (OR 0.735, 95% CI 0.699–0.773; p < 0.001). Consistent with these results, a recent case–control study involving 400 PCOS women compared with 480 healthy controls confirmed the reduced risk among G carriers of developing PCOS syndrome [117]. The mechanism by which this polymorphism could promote PCOS is still not understood. A recent study suggested that LHCGR rs13405728 could modulate the STON1 and FSHR transduction, thereby influencing metabolic processes and androgen receptor expression [118].

LHCGR missense polymorphisms

Finally, LHCGR missense polymorphism Asn312Ser (rs2293275) has been found to be associated with PCOS [93, 113, 119, 120]. In a meta-analysis collecting six association studies, Caucasian homozygous Ser carriers have an increased risk of developing ovarian hyperstimulation syndrome (OHSS) than Asn-homozygous and heterozygous (OR 4.11, CI 95% 1.03–16.38) [119]. Recently, a study involving 421 PCOS and 322 regularly menstruating women found the highest prevalence of the Ser variant among individuals with the disease [91].

Pharmacogenomic approach in women with clinical variants affecting the LH system

The pharmacogenomic approach consists of the prescription of medication based on the individual genetic profile. Specific genetic variants could influence the pharmacokinetics or pharmacodynamics of a drug. For instance, some individuals have increased or reduced receptor sensitivity to an exogenous medication or display a different drug half-life. In patients at risk of adverse events, the pharmacogenomic approach could help to minimize safety issues [121]. In the context of assisted reproduction, we have at least two possible adverse events linked to exogenous gonadotropin administration. One is OHSS, potentially a life-threatening event, in which an exaggerated ovarian response to gonadotropins occurs, leading to the development of multiple follicles, ascites, and thrombotic events [122, 123]. Moreover, women could have poor ovarian response to gonadotropins, leading to poor outcomes with assisted reproduction techniques and dropout from IVF treatment [124].

So far, very few studies have been conducted using a pharmacogenomic approach in the context of IVF. Regarding SNPs modulating LH/LHCGR signals, a prospective analysis of 193 women with a history of unsuccessful IVF cycles was performed [14]. Seventy-eight women were supplemented with 75 UI of LH from day 6 (control group), while 115 were supplemented with LH, according to LHCGR Asn312Ser phenotype, from day 1 (study group). Homozygous Asn carriers received no LH; 37.5 UI of LH was prescribed to heterozygous, and 75 UI to Ser-homozygous, carriers. Women receiving genotype-based LH personalized treatment had a higher clinical pregnancy rate than the control group (56/115 vs. 26/78; p = 0.049). However, limitations of this study must be acknowledged, such as its retrospective design and the duration of LH treatment, which was significantly higher in the study group than in controls. In another retrospective analysis, 533 women underwent a long protocol with FSH and LH coadministration, according to previously established criteria [125]. Authors found that Ser-homozygous women required more LH during COS, although they had a higher rate of pregnancies than women with other haplotypes [15].

Knowledge gaps and future research

Pharmacogenomics aims to improve the efficacy of therapeutic approaches, based on the susceptibility of genetic profiles. However, what is still debated is whether to promote the widespread use of this genetic information in clinical practice, since only a limited set of genetic variations have significant impact.

In specific subsets of ART patients, the reproductive outcome may benefit from LH supplementation. These patients consist in women with hypo-response to exogenous FSH alone [126,127,128,129], and women of advanced reproductive age (≥ 35 years) [130]. The genetic aspects beyond LH deficiency have been poorly understood. As previously reported, serine carriers of the LHCGR variant (rs2293275) may require higher amounts of LH, as a FSH supplement during COS, than other LHCGR genotypes [15]. Moreover carriers of LHB variant might need increased FSH dosage during COS [100]. The impact of LHCGR variant on pregnancy and cumulative pregnancy was observed by just one research group [83, 84] but unfortunately not corroborated by other studies [110, 111]. These mixed findings could be explained by inter-study differences in design and protocols. Another issue concerning pregnancy rate resides in the difficulty to reach the adequate sample size in IVF studies [131]. Finally, in our opinion the interaction among polymorphisms is still under-investigated. In other words, instead of focusing on single genetic association, future studies should focus more on simultaneous analysis of the genetic variants involved in COS [110, 132, 133].

So far, the lack of studies on gonadotropin polymorphisms could be explained by high costs of genetic tests, in comparison to hormonal assays, which are easier to perform, less expensive, and routinely used in clinical practice to guide COS during ART.

Dose-finding procedures, appropriate protocols, duration of stimulation, and number of cycles all warrant further investigation to improve our knowledge of ovarian response. Hence, the identification of genetic determinants for reduced ovarian response might lead to tailored medical approaches, reduce the overall costs of IVF treatments, and improve COS efficiency.

Predictive medicine also takes advantage of indexes to assess specific conditions. In ART, parameters such as the Follicle to Oocyte Index (FOI) and Follicular Output Rate (FORT) have been applied to practically define the ovarian response to COS [134]. However, no studies have been performed to evaluate the association between specific gonadotropin polymorphisms and ovarian sensitivity indexes. This knowledge might play a key role in increasing the chance of recruiting more oocytes and optimizing the chance of a live birth [135]. Informative results on whether such genetic polymorphisms could be associated with reduced response to COS may be obtained from poor responder women, who belong to groups 3 and 4 of the POSEIDON classification. Nonetheless, there is a need for large multicenter studies of real-world data, across different ages and ethnic groups, to further evaluate benefits of genetic testing in ART.

Conclusion

The pharmacogenomic approach based on LHB and/or LHCGR genotypes remains under-researched. The most promising SNP that might be used to personalize COS is the LHCGR Asn312Ser, but the evidence so far is poor and limited to very few studies. Although a possible cumulative effect between LH and LHCGR SNPs has been reported in several studies, these findings have yet to be supported by any randomized clinical trials. Without such investigations, it is not yet possible to suggest a pharmacogenomic approach in clinical practice.

Data availability

Not applicable.

Abbreviations

AKT:

Protein kinase B

AMP:

Adenosine monophosphate

ART:

Assisted reproductive therapy

Ca2+ :

Calcium ion

cAMP:

Cyclic adenosine monophosphate

CGA:

Human α subunit-coding gene

CGB:

Human β subunit-coding gene

COS:

Controlled ovarian stimulation

CREB:

cAMP response element–binding protein

ERK1/2:

Extracellular-regulated kinase 1/2

FSH[R]:

Follicle-stimulating hormone [receptor

GnRH:

Gonadotropin-releasing hormone

GPCR:

G protein-coupled receptor

hCG:

Human chorionic gonadotropin

IP3 :

Inositol trisphosphate

IVF:

in vitro fertilization

LH:

Luteinizing hormone

LHCGR:

Luteinizing hormone/choriogonadotropin receptor

LuRKO:

LH receptor knockout

OHSS:

Ovarian hyperstimulation syndrome

OR:

Odds ratio

PCOS:

Polycystic ovary syndrome

PKA:

Protein kinase A

PLC:

Phospholipase C

SNP:

Single-nucleotide polymorphism

TSH:

Thyroid-stimulating hormone

References

  1. Ascoli M, Fanelli F, Segaloff DL. The lutropin/choriogonadotropin receptor, a 2002 perspective. Endocr Rev. 2002;23:141–74.

    Article  CAS  PubMed  Google Scholar 

  2. Pierce JG, Faith MR, Giudice LC, Reeve JR. Structure and structure–function relationships in glycoprotein hormones. Ciba Found Symp. 1976;41:225–50.

    CAS  PubMed  Google Scholar 

  3. Casarini L, Santi D, Brigante G, Simoni M. Two hormones for one receptor: evolution, biochemistry, actions, and pathophysiology of LH and hCG. Endocr Rev. 2018;39:549–92.

    Article  PubMed  Google Scholar 

  4. Hallast P, Nagirnaja L, Margus T, Laan M. Segmental duplications and gene conversion: human luteinizing hormone/chorionic gonadotropin beta gene cluster. Genome Res. 2005;15:1535–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Fiddes JC, Goodman HM. The cDNA for the beta-subunit of human chorionic gonadotropin suggests evolution of a gene by readthrough into the 3’-untranslated region. Nature. 1980;286:684–7.

    Article  CAS  PubMed  Google Scholar 

  6. De Leo V, Musacchio MC, Di Sabatino A, Tosti C, Morgante G, Petraglia F. Present and future of recombinant gonadotropins in reproductive medicine. Curr Pharm Biotechnol. 2012;13:379–91.

    Article  PubMed  Google Scholar 

  7. Rivero-Müller A, Huhtaniemi I. Genetic variants of gonadotrophins and their receptors: impact on the diagnosis and management of the infertile patient. Best Pract Res Clin Endocrinol Metab. 2022;36:101596.

    Article  PubMed  Google Scholar 

  8. Conforti A, Vaiarelli A, Cimadomo D, Bagnulo F, Peluso S, Carbone L, et al. Pharmacogenetics of FSH action in the female. Front Endocrinol (Lausanne). 2019;10:398.

    Article  PubMed  Google Scholar 

  9. Lévy DP, Navarro JM, Schattman GL, Davis OK, Rosenwaks Z. The role of LH in ovarian stimulation: exogenous LH: let’s design the future. Hum Reprod. 2000;15:2258–65.

    Article  PubMed  Google Scholar 

  10. Alviggi C, Clarizia R, Mollo A, Ranieri A, De Placido G. Who needs LH in ovarian stimulation? Reprod Biomed Online. 2011;22:s33-41.

    Article  PubMed  Google Scholar 

  11. Humaidan P, Alviggi C, Fischer R, Esteves SC. The novel POSEIDON stratification of low prognosis patients in assisted reproductive technology and its proposed marker of successful outcome. F1000Res. 2016;5:2911.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Sunkara SK, Ramaraju GA, Kamath MS. Management strategies for POSEIDON group 2. Front Endocrinol (Lausanne). 2020;11:105.

    Article  PubMed  Google Scholar 

  13. Haahr T, Dosouto C, Alviggi C, Esteves SC, Humaidan P. Management strategies for POSEIDON groups 3 and 4. Front Endocrinol (Lausanne). 2019;10:614.

    Article  PubMed  Google Scholar 

  14. Ramaraju GA, Cheemakurthi R, Kalagara M, Prathigudupu K, Balabomma KL, Mahapatro P, et al. Effect of LHCGR gene polymorphism (rs2293275) on LH supplementation protocol outcomes in second IVF cycles: a retrospective study. Front Endocrinol (Lausanne). 2021;12:628169.

    Article  Google Scholar 

  15. Ramaraju GA, Cheemakurthi R, Prathigudupu K, Balabomma KL, Kalagara M, Thota S, et al. Role of Lh polymorphisms and r-hLh supplementation in GnRh agonist treated ART cycles: a cross sectional study. Eur J Obstet Gynecol Reprod Biol. 2018;222:119–25.

    Article  Google Scholar 

  16. Alviggi C, Pettersson K, Longobardi S, Andersen CY, Conforti A, De Rosa P, et al. A common polymorphic allele of the LH beta-subunit gene is associated with higher exogenous FSH consumption during controlled ovarian stimulation for assisted reproductive technology. Reprod Biol Endocrinol. 2013;11:51–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Duan J, Xu P, Cheng X, Mao C, Croll T, He X, et al. Structures of full-length glycoprotein hormone receptor signalling complexes. Nature. 2021;598:688–92.

    Article  PubMed  Google Scholar 

  18. Jagiello G, Ducayen M, Miller W, Graffeo J, Fang JS. Stimulation and inhibition with LH and other hormones of female mammalian meiosis in vitro. J Reprod Fertil. 1975;43:9–22.

    Article  CAS  PubMed  Google Scholar 

  19. Papadopoulos V, Carreau S, Drosdowsky MA. Effect of phorbol ester and phospholipase C on LH-stimulated steroidogenesis in purified rat leydig cells. FEBS Lett. 1985;188:312–6.

    Article  CAS  PubMed  Google Scholar 

  20. Gudermann T, Nichols C, Levy FO, Birnbaumer M, Birnbaumer L. Ca2 + mobilization by the LH receptor expressed in Xenopus oocytes independent of 3’,5’-cyclic adenosine monophosphate formation: evidence for parallel activation of two signaling pathways. Mol Endocrinol. 1992;6:272–8.

    CAS  PubMed  Google Scholar 

  21. Montminy MR, Bilezikjian LM. Binding of a nuclear protein to the cyclic-AMP response element of the somatostatin gene. Nature. 1987;328:175–8.

    Article  CAS  PubMed  Google Scholar 

  22. Casarini L, Lazzaretti C, Paradiso E, Limoncella S, Riccetti L, Sperduti S, et al. Membrane estrogen receptor (GPER) and follicle-stimulating hormone receptor (FSHR) heteromeric complexes promote human ovarian follicle survival. iScience. 2020;23: 101812.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Casarini L, Reiter E, Simoni M. β-arrestins regulate gonadotropin receptor-mediated cell proliferation and apoptosis by controlling different FSHR or LHCGR intracellular signaling in the hGL5 cell line. Mol Cell Endocrinol. 2016;437:11–21.

    Article  CAS  PubMed  Google Scholar 

  24. Zwain IH, Amato P. cAMP-induced apoptosis in granulosa cells is associated with up-regulation of P53 and bax and down-regulation of clusterin. Endocr Res. 2001;27:233–49.

    Article  CAS  PubMed  Google Scholar 

  25. Regan SLP, Knight PG, Yovich JL, Leung Y, Arfuso F, Dharmarajan A. Granulosa cell apoptosis in the ovarian follicle – a changing view. Front Endocrinol (Lausanne). 2018;9:61.

    Article  PubMed  Google Scholar 

  26. Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev. 2011;32:81–151.

    Article  CAS  PubMed  Google Scholar 

  27. Tajima K, Yoshii K, Fukuda S, Orisaka M, Miyamoto K, Amsterdam A, et al. Luteinizing hormone-induced extracellular-signal regulated kinase activation differently modulates progesterone and androstenedione production in bovine theca cells. Endocrinology. 2005;146:2903–10.

    Article  CAS  PubMed  Google Scholar 

  28. Menon B, Franzo-Romain M, Damanpour S, Menon KMJ. Luteinizing hormone receptor mRNA down-regulation is mediated through ERK-dependent induction of RNA binding protein. Mol Endocrinol. 2011;25:282–90.

    Article  CAS  PubMed  Google Scholar 

  29. Ren X-R, Reiter E, Ahn S, Kim J, Chen W, Lefkowitz RJ. Different G protein-coupled receptor kinases govern G protein and beta-arrestin-mediated signaling of V2 vasopressin receptor. Proc Natl Acad Sci U S A. 2005;102:1448–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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;45:101–8.

    Article  CAS  PubMed  Google Scholar 

  31. Tohgo A, Choy EW, Gesty-Palmer D, Pierce KL, Laporte S, Oakley RH, et al. The stability of the G protein-coupled receptor-beta-arrestin interaction determines the mechanism and functional consequence of ERK activation. J Biol Chem. 2003;278:6258–67.

    Article  CAS  PubMed  Google Scholar 

  32. Mertens-Walker I, Bolitho C, Baxter RC, Marsh DJ. Gonadotropin-induced ovarian cancer cell migration and proliferation require extracellular signal-regulated kinase 1/2 activation regulated by calcium and protein kinase C{delta}. Endocr Relat Cancer. 2010;17:335–49.

    Article  CAS  PubMed  Google Scholar 

  33. Manna PR, Pakarinen P, El-Hefnawy T, Huhtaniemi IT. Functional assessment of the calcium messenger system in cultured mouse Leydig tumor cells: regulation of human chorionic gonadotropin-induced expression of the steroidogenic acute regulatory protein. Endocrinology. 1999;140:1739–51.

    Article  CAS  PubMed  Google Scholar 

  34. Pezzi V, Clark BJ, Ando S, Stocco DM, Rainey WE. Role of calmodulin-dependent protein kinase II in the acute stimulation of aldosterone production. J Steroid Biochem Mol Biol. 1996;58:417–24.

    Article  CAS  PubMed  Google Scholar 

  35. Zhu X, Gilbert S, Birnbaumer M, Birnbaumer L. Dual signaling potential is common among Gs-coupled receptors and dependent on receptor density. Mol Pharmacol. 1994;46:460–9.

    Article  CAS  PubMed  Google Scholar 

  36. Tai P, Shiraishi K, Ascoli M. Activation of the lutropin/choriogonadotropin receptor inhibits apoptosis of immature Leydig cells in primary culture. Endocrinology. 2009;150:3766–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Andric N, Thomas M, Ascoli M. Transactivation of the epidermal growth factor receptor is involved in the lutropin receptor-mediated down-regulation of ovarian aromatase expression in vivo. Mol Endocrinol. 2010;24:552–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Light A, Hammes SR. Membrane receptor cross talk in steroidogenesis: recent insights and clinical implications. Steroids. 2013;78:633–8.

    Article  CAS  PubMed  Google Scholar 

  39. Casarini L, Santi D, Simoni M, Potì F. Spare luteinizing hormone receptors: facts and fiction. Trends Endocrinol Metab. 2018;29:208–17.

    Article  CAS  PubMed  Google Scholar 

  40. Ji I, Lee C, Jeoung M, Koo Y, Sievert GA, Ji TH. Trans-activation of mutant follicle-stimulating hormone receptors selectively generates only one of two hormone signals. Mol Endocrinol. 2004;18:968–78.

    Article  CAS  PubMed  Google Scholar 

  41. Feng X, Zhang M, Guan R, Segaloff DL. Heterodimerization between the lutropin and follitropin receptors is associated with an attenuation of hormone-dependent signaling. Endocrinology. 2013;154:3925–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tranchant T, Durand G, Gauthier C, Crépieux P, Ulloa-Aguirre A, Royère D, et al. Preferential β-arrestin signalling at low receptor density revealed by functional characterization of the human FSH receptor A189 V mutation. Mol Cell Endocrinol. 2011;331:109–18.

    Article  CAS  PubMed  Google Scholar 

  43. Donadeu FX, Ascoli M. The differential effects of the gonadotropin receptors on aromatase expression in primary cultures of immature rat granulosa cells are highly dependent on the density of receptors expressed and the activation of the inositol phosphate cascade. Endocrinology. 2005;146:3907–16.

    Article  CAS  PubMed  Google Scholar 

  44. Jeppesen JV, Kristensen SG, Nielsen ME, Humaidan P, Dal Canto M, Fadini R, et al. LH-receptor gene expression in human granulosa and cumulus cells from antral and preovulatory follicles. J Clin Endocrinol Metab. 2012;97:E1524-1531.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hunzicker-Dunn M, Barisas G, Song J, Roess DA. Membrane organization of luteinizing hormone receptors differs between actively signaling and desensitized receptors. J Biol Chem. 2003;278:42744–9.

    Article  CAS  PubMed  Google Scholar 

  46. Hirakawa T, Galet C, Kishi M, Ascoli M. GIPC binds to the human lutropin receptor (hLHR) through an unusual PDZ domain binding motif, and it regulates the sorting of the internalized human choriogonadotropin and the density of cell surface hLHR. J Biol Chem. 2003;278:49348–57.

    Article  CAS  PubMed  Google Scholar 

  47. Hanyaloglu AC. Advances in membrane trafficking and endosomal signaling of G protein-coupled receptors. Int Rev Cell Mol Biol. 2018;339:93–131.

    Article  CAS  PubMed  Google Scholar 

  48. Thomsen ARB, Plouffe B, Cahill TJ, Shukla AK, Tarrasch JT, Dosey AM, et al. GPCR-G protein-β-arrestin super-complex mediates sustained G protein signaling. Cell. 2016;166:907–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sposini S, Jean-Alphonse FG, Ayoub MA, Oqua A, West C, Lavery S, et al. Integration of GPCR signaling and sorting from very early endosomes via opposing APPL1 mechanisms. Cell Rep. 2017;21:2855–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Casarini L, Lispi M, Longobardi S, Milosa F, La Marca A, Tagliasacchi D, et al. LH and hCG action on the same receptor results in quantitatively and qualitatively different intracellular signalling. PLoS One. 2012;7:e46682.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Casarini L, Riccetti L, De Pascali F, Nicoli A, Tagliavini S, Trenti T, et al. Follicle-stimulating hormone potentiates the steroidogenic activity of chorionic gonadotropin and the anti-apoptotic activity of luteinizing hormone in human granulosa-lutein cells in vitro. Mol Cell Endocrinol. 2016;422:103–14.

    Article  CAS  PubMed  Google Scholar 

  52. Riccetti L, De Pascali F, Gilioli L, Potì F, Giva LB, Marino M, et al. Human LH and hCG stimulate differently the early signalling pathways but result in equal testosterone synthesis in mouse leydig cells in vitro. Reprod Biol Endocrinol. 2017;15:2.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Riccetti L, Yvinec R, Klett D, Gallay N, Combarnous Y, Reiter E, et al. Human luteinizing hormone and chorionic gonadotropin display biased agonism at the LH and LH/CG receptors. Sci Rep. 2017;7:940.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Casarini L, Riccetti L, De Pascali F, Gilioli L, Marino M, Vecchi E, et al. Estrogen modulates specific life and death signals induced by LH and hCG in human primary granulosa cells in vitro. Int J Mol Sci. 2017;18:926.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Santi D, Casarini L, Alviggi C, Simoni M. Efficacy of follicle-stimulating hormone (FSH) alone, FSH + luteinizing hormone, human menopausal gonadotropin or FSH + human chorionic gonadotropin on assisted reproductive technology outcomes in the personalized medicine era: a meta-analysis. Front Endocrinol. 2017;8:114.

    Article  Google Scholar 

  56. Rulli SB, Kuorelahti A, Karaer O, Pelliniemi LJ, Poutanen M, Huhtaniemi I. Reproductive disturbances, pituitary lactotrope adenomas, and mammary gland tumors in transgenic female mice producing high levels of human chorionic gonadotropin. Endocrinology. 2002;143:4084–95.

    Article  CAS  PubMed  Google Scholar 

  57. Peltoketo H, Rivero-Müller A, Ahtiainen P, Poutanen M, Huhtaniemi I. Consequences of genetic manipulations of gonadotrophins and gonadotrophin receptors in mice. Ann Endocrinol (Paris). 2010;71:170–6.

    Article  CAS  PubMed  Google Scholar 

  58. Risma KA, Clay CM, Nett TM, Wagner T, Yun J, Nilson JH. Targeted overexpression of luteinizing hormone in transgenic mice leads to infertility, polycystic ovaries, and ovarian tumors. Proc Natl Acad Sci U S A. 1995;92:1322–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Kendall SK, Samuelson LC, Saunders TL, Wood RI, Camper SA. Targeted disruption of the pituitary glycoprotein hormone alpha-subunit produces hypogonadal and hypothyroid mice. Genes Dev. 1995;9:2007–19.

    Article  CAS  PubMed  Google Scholar 

  60. Ma X, Dong Y, Matzuk MM, Kumar TR. Targeted disruption of luteinizing hormone beta-subunit leads to hypogonadism, defects in gonadal steroidogenesis, and infertility. Proc Natl Acad Sci U S A. 2004;101:17294–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. O’Shaughnessy PJ, Fleming LM, Jackson G, Hochgeschwender U, Reed P, Baker PJ. Adrenocorticotropic hormone directly stimulates testosterone production by the fetal and neonatal mouse testis. Endocrinology. 2003;144:3279–84.

    Article  PubMed  Google Scholar 

  62. El-Gehani F, Zhang FP, Pakarinen P, Rannikko A, Huhtaniemi I. Gonadotropin-independent regulation of steroidogenesis in the fetal rat testis. Biol Reprod. 1998;58:116–23.

    Article  CAS  PubMed  Google Scholar 

  63. Zhang F-P, Pakarainen T, Poutanen M, Toppari J, Huhtaniemi I. The low gonadotropin-independent constitutive production of testicular testosterone is sufficient to maintain spermatogenesis. Proc Natl Acad Sci U S A. 2003;100:13692–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oduwole OO, Peltoketo H, Poliandri A, Vengadabady L, Chrusciel M, Doroszko M, et al. Constitutively active follicle-stimulating hormone receptor enables androgen-independent spermatogenesis. J Clin Invest. 2018;128:1787–92.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Jonas KC, Rivero Müller A, Oduwole O, Peltoketo H, Huhtaniemi I. The luteinizing hormone receptor knockout mouse as a tool to probe the in vivo actions of gonadotropic hormones/receptors in females. Endocrinology. 2021;162:bqab035.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Rull K, Laan M. Expression of beta-subunit of HCG genes during normal and failed pregnancy. Hum Reprod. 2005;20:3360–8.

    Article  CAS  PubMed  Google Scholar 

  67. Conforti A, Tüttelmann F, Alviggi C, Behre HM, Fischer R, Hu L, et al. Effect of genetic variants of gonadotropins and their receptors on ovarian stimulation outcomes: a Delphi consensus. Front Endocrinol (Lausanne). 2021;12:797365.

    Article  PubMed  Google Scholar 

  68. Pettersson K, Ding YQ, Huhtaniemi I. An immunologically anomalous luteinizing hormone variant in a healthy woman. J Clin Endocrinol Metab. 1992;74:164–71.

    CAS  PubMed  Google Scholar 

  69. Nilsson C, Jiang M, Pettersson K, Iitiä A, Mäkelä M, Simonsen H, et al. Determination of a common genetic variant of luteinizing hormone using DNA hybridization and immunoassays. Clin Endocrinol (Oxf). 1998;49:369–76.

    Article  CAS  PubMed  Google Scholar 

  70. Suganuma N, Furui K, Kikkawa F, Tomoda Y, Furuhashi M. Effects of the mutations (Trp8 --> arg and Ile15 --> thr) in human luteinizing hormone (LH) beta-subunit on LH bioactivity in vitro and in vivo. Endocrinology. 1996;137:831–8.

    Article  CAS  PubMed  Google Scholar 

  71. Jiang M, Lamminen T, Pakarinen P, Hellman J, Manna P, Herrera RJ, et al. A novel Ala(-3)thr mutation in the signal peptide of human luteinizing hormone beta-subunit: potentiation of the inositol phosphate signalling pathway and attenuation of the adenylate cyclase pathway by recombinant variant hormone. Mol Hum Reprod. 2002;8:201–12.

    Article  CAS  PubMed  Google Scholar 

  72. Liao WX, Goh HH, Roy AC. Functional characterization of a natural variant of luteinizing hormone. Hum Genet. 2002;111:219–24.

    Article  CAS  PubMed  Google Scholar 

  73. Liu S, Ogata T, Maruyama T, Yoshimura Y, Ishizuka B. Association of common LH variant with hyperfunctional promoter in a Japanese infertile woman. Endocr J. 2005;52:781–4.

    Article  CAS  PubMed  Google Scholar 

  74. Bosco L, Ruvolo G, Luparello C, Ferrari S, Valerio D, Santi D, et al. Gene expression and apoptosis levels in cumulus cells of patients with polymorphisms of FSHR and LHB undergoing in vitro fertilization program. Cell Physiol Biochem. 2017;43:2391–404.

    Article  CAS  PubMed  Google Scholar 

  75. Tapanainen JS, Koivunen R, Fauser BC, Taylor AE, Clayton RN, Rajkowa M, et al. A new contributing factor to polycystic ovary syndrome: the genetic variant of luteinizing hormone. J Clin Endocrinol Metab. 1999;84:1711–5.

    CAS  PubMed  Google Scholar 

  76. Branavan U, Muneeswaran K, Wijesundera S, Jayakody S, Chandrasekharan V, Wijeyaratne C. Identification of selected genetic polymorphisms in polycystic ovary syndrome in Sri Lankan women using low-cost genotyping techniques. PLoS One. 2018;13: e0209830.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Jiang M, Pakarinen P, Zhang FP, El-Hefnawy T, Koskimies P, Pettersson K, et al. A common polymorphic allele of the human luteinizing hormone beta-subunit gene: additional mutations and differential function of the promoter sequence. Hum Mol Genet. 1999;8:2037–46.

    Article  CAS  PubMed  Google Scholar 

  78. Punab AM, Grigorova M, Punab M, Adler M, Kuura T, Poolamets O, et al. Carriers of variant luteinizing hormone (V-LH) among 1593 baltic men have significantly higher serum LH. Andrology. 2015;3:512–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Lamminen T, Jiang M, Manna PR, Pakarinen P, Simonsen H, Herrera RJ, et al. Functional study of a recombinant form of human LH beta-subunit variant carrying the gly(102)ser mutation found in Asian populations. Mol Hum Reprod. 2002;8:887–92.

    Article  CAS  PubMed  Google Scholar 

  80. Zhao Y, Chen T, Zhou Y, Li K, Xiao J. An association study between the genetic polymorphisms within GnRHI, LHβ and FSHβ genes and central precocious puberty in Chinese girls. Neurosci Lett. 2010;486:188–92.

    Article  CAS  PubMed  Google Scholar 

  81. Dasgupta S, Sirisha PVS, Neelaveni K, Anuradha K, Sudhakar G, Reddy BM. Role of luteinizing hormone β-subunit gene variants among south Indian women with polycystic ovary syndrome. Gene. 2012;494:51–6.

    Article  CAS  PubMed  Google Scholar 

  82. Cho SG, Bhoumik A, Broday L, Ivanov V, Rosenstein B, Ronai Z. TIP49b, a regulator of activating transcription factor 2 response to stress and DNA damage. Mol Cell Biol. 2001;21:8398–413.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Lindgren I, Nenonen H, Henic E, Bungum L, Prahl A, Bungum M, et al. Gonadotropin receptor variants are linked to cumulative live birth rate after in vitro fertilization. J Assist Reprod Genet. 2019;36:29–38.

    Article  CAS  PubMed  Google Scholar 

  84. Lindgren I, Bååth M, Uvebrant K, Dejmek A, Kjaer L, Henic E, et al. Combined assessment of polymorphisms in the LHCGR and FSHR genes predict chance of pregnancy after in vitro fertilization. Hum Reprod. 2016;31:672–83.

    Article  CAS  PubMed  Google Scholar 

  85. Chen Z-J, Zhao H, He L, Shi Y, Qin Y, Shi Y, et al. Genome-wide association study identifies susceptibility loci for polycystic ovary syndrome on chromosome 2p16.3, 2p21 and 9q33.3. Nat Genet. 2011;43:55–9.

    Article  PubMed  Google Scholar 

  86. Casarini L, Brigante G. The polycystic ovary syndrome evolutionary paradox: a genome-wide association studies-based, in silico, evolutionary explanation. J Clin Endocrinol Metab. 2014;99:E2412-2420.

    Article  CAS  PubMed  Google Scholar 

  87. Shi Y, Zhao H, Shi Y, Cao Y, Yang D, Li Z, et al. Genome-wide association study identifies eight new risk loci for polycystic ovary syndrome. Nat Genet. 2012;44:1020–5.

    Article  CAS  PubMed  Google Scholar 

  88. Jones MR, Brower MA, Xu N, Cui J, Mengesha E, Chen Y-DI, et al. Systems genetics reveals the functional context of PCOS loci and identifies genetic and molecular mechanisms of disease heterogeneity. PLoS Genet. 2015;11:e1005455.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Deswal R, Nanda S, Dang AS. Association of luteinizing hormone and LH receptor gene polymorphism with susceptibility of polycystic ovary syndrome. Syst Biol Reprod Med. 2019;65:400–8.

    Article  CAS  PubMed  Google Scholar 

  90. Zhang Y-J, Li L, Wang Z-J, Zhang X-J, Zhao H, Zhao Y, et al. Association study between variants in LHCGR DENND1A and THADA with preeclampsia risk in Han Chinese populations. J Matern Fetal Neonatal Med. 2019;32:3801–5.

    Article  CAS  PubMed  Google Scholar 

  91. Singh S, Kaur M, Kaur R, Beri A, Kaur A. Association analysis of LHCGR variants and polycystic ovary syndrome in Punjab: a case-control approach. BMC Endocr Disord. 2022;22:335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Liaqat I, Jahan N, Krikun G, Taylor HS. Genetic polymorphisms in Pakistani women with polycystic ovary syndrome. Reprod Sci. 2015;22:347–57.

    Article  CAS  PubMed  Google Scholar 

  93. Bassiouny YA, Rabie WA, Hassan AA, Darwish RK. Association of the luteinizing hormone/choriogonadotropin receptor gene polymorphism with polycystic ovary syndrome. Gynecol Endocrinol. 2014;30:428–30.

    Article  CAS  PubMed  Google Scholar 

  94. Cui L, Zhao H, Zhang B, Qu Z, Liu J, Liang X, et al. Genotype–phenotype correlations of PCOS susceptibility SNPs identified by GWAS in a large cohort of Han Chinese women. Hum Reprod. 2013;28:538–44.

    Article  CAS  PubMed  Google Scholar 

  95. Liu N, Ma Y, Wang S, Zhang X, Zhang Q, Zhang X, et al. Association of the genetic variants of luteinizing hormone, luteinizing hormone receptor and polycystic ovary syndrome. Reprod Biol Endocrinol. 2012;10: 36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Lidaka L, Bekere L, Rota A, Isakova J, Lazdane G, Kivite-Urtane A, et al. Role of single nucleotide variants in FSHR, GNRHR, ESR2 and LHCGR genes in adolescents with polycystic ovary syndrome. Diagnostics (Basel). 2021;11:2327.

    Article  CAS  PubMed  Google Scholar 

  97. Makhdoomi MJ, Shah IA, Rashid R, Rashid A, Singh S, Shah ZA, et al. Effect modification of LHCGR gene variant (rs2293275) on clinico-biochemical profile, and levels of luteinizing hormone in polycystic ovary syndrome patients. Biochem Genet. 2023;61:1418–32.

    Article  CAS  PubMed  Google Scholar 

  98. Simoni M, Tüttelmann F, Michel C, Böckenfeld Y, Nieschlag E, Gromoll J. Polymorphisms of the luteinizing hormone/chorionic gonadotropin receptor gene: association with maldescended testes and male infertility. Pharmacogenet Genomics. 2008;18:193–200.

    Article  CAS  PubMed  Google Scholar 

  99. Singh R, Kaur S, Yadav S, Bhatia S. Gonadotropins as pharmacological agents in assisted reproductive technology and polycystic ovary syndrome. Trends Endocrinol Metab. 2023;S1043–2760(23):00031.

    Google Scholar 

  100. Alviggi C, Clarizia R, Pettersson K, Mollo A, Humaidan P, Strina I, et al. Suboptimal response to GnRHa long protocol is associated with a common LH polymorphism. Reprod Biomed Online. 2011;22(Suppl 1):S67-72.

    Article  PubMed  Google Scholar 

  101. Ku Y, Hong MA, Chae SJ, Lim KS, Lee W-D, Lim JH, et al. The effects of luteinising hormone gene polymorphism on the outcomes of in vitro fertilisation and embryo transfer. J Obstet Gynaecol. 2021;41:1092–6.

    Article  CAS  PubMed  Google Scholar 

  102. Garcia-Velasco JA, Coelingh Bennink HJT, Epifanio R, Escudero E, Pellicer A, Simón C. High-dose recombinant LH add-back strategy using high-dose GnRH antagonist is an innovative protocol compared with standard GnRH antagonist. Reprod Biomed Online. 2007;15:280–7.

    Article  CAS  PubMed  Google Scholar 

  103. Kol S. LH supplementation in ovarian stimulation for IVF: the individual, LH deficient, patient perspective. Gynecol Obstet Invest. 2020;85:307–11.

    Article  CAS  PubMed  Google Scholar 

  104. Barmat LI, Chantilis SJ, Hurst BS, Dickey RP. A randomized prospective trial comparing gonadotropin-releasing hormone (GnRH) antagonist/recombinant follicle-stimulating hormone (rFSH) versus GnRH-agonist/rFSH in women pretreated with oral contraceptives before in vitro fertilization. Fertil Steril. 2005;83:321–30.

    Article  CAS  PubMed  Google Scholar 

  105. Liao WX, Roy AC, Chan C, Arulkumaran S, Ratnam SS. A new molecular variant of luteinizing hormone associated with female infertility. Fertil Steril. 1998;69:102–6.

    Article  CAS  PubMed  Google Scholar 

  106. Davar R, Tabibnejad N, Kalantar SM, Sheikhha MH. The luteinizing hormone beta-subunit exon 3 (Gly102Ser) gene mutation and ovarian responses to controlled ovarian hyperstimulation. Iran J Reprod Med. 2014;12:667–72.

    PubMed  PubMed Central  Google Scholar 

  107. Gardner DK, Lane M, Schoolcraft WB. Physiology and culture of the human blastocyst. J Reprod Immunol. 2002;55:85–100.

    Article  CAS  PubMed  Google Scholar 

  108. Gardner DK, Lane M, Stevens J, Schlenker T, Schoolcraft WB. Blastocyst score affects implantation and pregnancy outcome: towards a single blastocyst transfer. Fertil Steril. 2000;73:1155–8.

    Article  CAS  PubMed  Google Scholar 

  109. Guo C, Yu H, Feng G, Lv Q, Liu X, Liu X. Associations of FSHR and LHCGR gene variants with ovarian reserve and clinical pregnancy rates. Reprod Biomed Online. 2021;43:561–9.

    Article  CAS  PubMed  Google Scholar 

  110. Alviggi C, Longobardi S, Papaleo E, Santi D, Alfano S, Vanni VS, et al. Genetic variants of gonadotropins and their receptors could influence controlled ovarian stimulation: IVF data from a prospective multicenter study. Genes (Basel). 2023;14:1269.

    Article  CAS  PubMed  Google Scholar 

  111. Pirtea P, de Ziegler D, Marin D, Sun L, Tao X, Ayoubi JM, et al. Gonadotropin receptor polymorphisms (FSHR N680S and LHCGR N312S) are not predictive of clinical outcome and live birth in assisted reproductive technology. Fertil Steril. 2022;118:494–503.

    Article  CAS  PubMed  Google Scholar 

  112. Piersma D, Verhoef-Post M, Look MP, Uitterlinden AG, Pols HAP, Berns EMJJ, et al. Polymorphic variations in exon 10 of the luteinizing hormone receptor: functional consequences and associations with breast cancer. Mol Cell Endocrinol. 2007;276:63–70.

    Article  CAS  PubMed  Google Scholar 

  113. Capalbo A, Sagnella F, Apa R, Fulghesu AM, Lanzone A, Morciano A, et al. The 312 N variant of the luteinizing hormone/choriogonadotropin receptor gene (LHCGR) confers up to 2·7-fold increased risk of polycystic ovary syndrome in a sardinian population. Clin Endocrinol (Oxf). 2012;77:113–9.

    Article  CAS  PubMed  Google Scholar 

  114. Valkenburg O, Uitterlinden AG, Piersma D, Hofman A, Themmen APN, de Jong FH, et al. Genetic polymorphisms of GnRH and gonadotrophic hormone receptors affect the phenotype of polycystic ovary syndrome. Hum Reprod. 2009;24:2014–22.

    Article  CAS  PubMed  Google Scholar 

  115. Aschim EL, Oldenburg J, Kristiansen W, Giwercman A, Witczak O, Fosså SD, et al. Genetic variations associated with the effect of testicular cancer treatment on gonadal hormones. Hum Reprod. 2014;29:2844–51.

    Article  CAS  PubMed  Google Scholar 

  116. Alviggi C, Conforti A, Cariati F, Alfano S, Strina I, Huhtaniemi I, et al. Impact of polymorphisms of gonadotropins and their receptors on controlled ovarian stimulation: a prospective observational study. Hum Reprod. 2016;31:443–443.

    Google Scholar 

  117. Wan P, Meng L, Huang C, Dai B, Jin Y, Chai L, et al. Replication study and meta-analysis of selected genetic variants and polycystic ovary syndrome susceptibility in Asian population. J Assist Reprod Genet. 2021;38:2781–9.

    Article  PubMed  PubMed Central  Google Scholar 

  118. Cao CH, Wei Y, Liu R, Lin XR, Luo JQ, Zhang QJ, et al. Three-dimensional genome interactions identify potential adipocyte metabolism-associated gene STON1 and immune-correlated gene FSHR at the rs13405728 locus in polycystic ovary syndrome. Front Endocrinol (Lausanne). 2021;12:686054.

    Article  PubMed  Google Scholar 

  119. Zou J, Wu D, Liu Y, Tan S. Association of luteinizing hormone/choriogonadotropin receptor gene polymorphisms with polycystic ovary syndrome risk: a meta-analysis. Gynecol Endocrinol. 2019;35:81–5.

    Article  CAS  PubMed  Google Scholar 

  120. Thathapudi S, Kodati V, Erukkambattu J, Addepally U, Qurratulain H. Association of luteinizing hormone chorionic gonadotropin receptor gene polymorphism (rs2293275) with polycystic ovarian syndrome. Genet Test Mol Biomarkers. 2015;19:128–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Sychev DA, Malova EU. Evidence-based pharmacogenetics: is it possible? Int J Risk Saf Med. 2015;27(Suppl 1):S97-98.

    Article  PubMed  Google Scholar 

  122. Iorio GG, Rovetto MY, Conforti A, Carbone L, Vallone R, Cariati F, et al. Severe ovarian hyperstimulation syndrome in a woman with breast cancer under letrozole triggered with GnRH agonist: a case report and review of the literature. Front Reprod Health. 2021;3: 704153.

    Article  PubMed  PubMed Central  Google Scholar 

  123. Feferkorn I, Ata B, Esteves SC, La Marca A, Paulson R, Blockeel C, et al. The HERA (Hyper-response risk Assessment) Delphi consensus definition of hyper-responders for in-vitro fertilization. J Assist Reprod Genet. 2023;40:1071–81.

    Article  PubMed  PubMed Central  Google Scholar 

  124. Verberg MFG, Eijkemans MJC, Heijnen EMEW, Broekmans FJ, de Klerk C, Fauser BCJM, et al. Why do couples drop-out from IVF treatment? A prospective cohort study. Hum Reprod. 2008;23:2050–5.

    Article  CAS  PubMed  Google Scholar 

  125. Wong PC, Qiao J, Ho C, Ramaraju GA, Wiweko B, Takehara Y, et al. Current opinion on use of luteinizing hormone supplementation in assisted reproduction therapy: an Asian perspective. Reprod Biomed Online. 2011;23:81–90.

    Article  CAS  PubMed  Google Scholar 

  126. De Placido G, Alviggi C, Perino A, Strina I, Lisi F, Fasolino A, et al. Recombinant human LH supplementation versus recombinant human FSH (rFSH) step-up protocol during controlled ovarian stimulation in normogonadotrophic women with initial inadequate ovarian response to rFSH. A multicentre, prospective, randomized controlled trial. Hum Reprod. 2005;20:390–6.

    Article  PubMed  Google Scholar 

  127. Ferraretti AP, Gianaroli L, Magli MC, D’angelo A, Farfalli V, Montanaro N. Exogenous luteinizing hormone in controlled ovarian hyperstimulation for assisted reproduction techniques. Fertil Steril. 2004;82:1521–6.

    Article  CAS  PubMed  Google Scholar 

  128. Lisi F, Rinaldi L, Fishel S, Caserta D, Lisi R, Campbell A. Evaluation of two doses of recombinant luteinizing hormone supplementation in an unselected group of women undergoing follicular stimulation for in vitro fertilization. Fertil Steril. 2005;83:309–15.

    Article  CAS  PubMed  Google Scholar 

  129. Ruvolo G, Bosco L, Pane A, Morici G, Cittadini E, Roccheri MC. Lower apoptosis rate in human cumulus cells after administration of recombinant luteinizing hormone to women undergoing ovarian stimulation for in vitro fertilization procedures. Fertil Steril. 2007;87:542–6.

    Article  CAS  PubMed  Google Scholar 

  130. Conforti A, Esteves SC, Humaidan P, Longobardi S, D’Hooghe T, Orvieto R, et al. Recombinant human luteinizing hormone co-treatment in ovarian stimulation for assisted reproductive technology in women of advanced reproductive age: a systematic review and meta-analysis of randomized controlled trials. Reprod Biol Endocrinol. 2021;19:91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Clarke JF, van Rumste MME, Farquhar CM, Johnson NP, Mol BWJ, Herbison P. Measuring outcomes in fertility trials: can we rely on clinical pregnancy rates? Fertil Steril. 2010;94:1647–51.

    Article  PubMed  Google Scholar 

  132. Desai SS, Achrekar SK, Paranjape SR, Desai SK, Mangoli VS, Mahale SD. Association of allelic combinations of FSHR gene polymorphisms with ovarian response. Reprod Biomed Online. 2013;27:400–6.

    Article  CAS  PubMed  Google Scholar 

  133. Allegra A, Marino A, Raimondo S, Maiorana A, Gullo S, Scaglione P, et al. The carriers of the A/G-G/G allelic combination of the c.2039 a > G and c.-29 G > A FSH receptor polymorphisms retrieve the highest number of oocytes in IVF/ICSI cycles. J Assist Reprod Genet. 2017;34:263–73.

    Article  PubMed  Google Scholar 

  134. Alviggi C, Conforti A, Esteves SC, Vallone R, Venturella R, Staiano S, et al. Understanding ovarian hypo-response to exogenous gonadotropin in ovarian stimulation and its new proposed marker-the Follicle-To-Oocyte (FOI) index. Front Endocrinol (Lausanne). 2018;9:589.

    Article  PubMed  Google Scholar 

  135. Bosch E, Alviggi C, Lispi M, Conforti A, Hanyaloglu AC, Chuderland D, et al. Reduced FSH and LH action: implications for medically assisted reproduction. Hum Reprod. 2021;36:1469–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by Le Studium Loire Valley Institute for Advanced Studies, Orléans, and Tours, France, through the ARD CVL Biopharmaceuticals program funded by the Centre-Val de Loire region.

About this supplement

This article has been published as part of Reproductive Biology and Endocrinology, Volume 23 Supplement 01, 2025:Luteinizing Hormone throughout the fertility journey. The full contents of the supplement are available at https://biomedcentral-rbej.publicaciones.saludcastillayleon.es/articles/supplements/volume-23-supplement-1.

Funding

The authors received no specific funding for this work.

Author information

Authors and Affiliations

Authors

Contributions

All authors (AC, RDG, MG, CA, and LC) were involved in the drafting, review, and final approval of the manuscript for publication.

Corresponding author

Correspondence to Alessandro Conforti.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary Information

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Conforti, A., Di Girolamo, R., Guida, M. et al. Pharmacogenomic of LH and its receptor: are we ready for clinical practice?. Reprod Biol Endocrinol 23 (Suppl 1), 29 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-025-01359-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12958-025-01359-2

Keywords