Skip to main content

Identifying therapeutic targets for primary ovarian insufficiency through integrated genomic analyses

Abstract

Background

Primary ovarian insufficiency (POI) is a disorder characterized by the premature decline in ovarian function, leading to significant fertility and health impacts on women under 40. The unclear etiology of POI hinders the development of effective treatments, highlighting the need for novel therapeutic targets.

Methods

This study employed genome-wide association analysis (GWAS) integrated with expression quantitative trait loci (eQTL) data from the GTEx and eQTLGen databases. Mendelian randomization (MR) and colocalization analyses were conducted to investigate causal relationships between genetic variants and POI and to identify potential therapeutic targets.

Results

We identified 431 genes with available index cis-eQTL signals, of which four (HM13, FANCE, RAB2A, and MLLT10) were significantly associated with POI. Colocalization analysis revealed strong evidence for FANCE and RAB2A, indicating their potential as therapeutic targets. Subsequent druggability assessments identified FANCE and RAB2A as promising candidates for POI treatment, supported by their involvement in DNA repair and autophagy regulation, respectively.

Conclusions

Our study establishes a causal link between specific genes and POI, highlighting FANCE and RAB2A as potential drug targets. These findings provide a foundation for future research and therapeutic development, aiming to improve outcomes for women with POI. Validation in further trials is necessary to confirm these potential targets.

Background

Primary ovarian insufficiency (POI) is a disorder characterized by a decline in ovarian function, loss of oocytes and folliculogenesis, and elevated gonadotropin levels, typically occurring in women under 40 years of age [1]. POI significantly affects women’s fertility and quality of life [2]. The global prevalence of POI is approximately 3.7% and is increasing [3]. POI results from a premature decrease in ovarian follicles, accelerated follicle destruction, or a poor follicular response to gonadotropins. The pathogenesis remains inconclusive, with various factors implicated, including genetic, autoimmune, toxic, metabolic, infectious, and iatrogenic factors [1]. Hormone replacement therapy (HRT) is the primary pharmacological treatment for POI, yet scientific evidence supporting its efficacy in restoring ovarian function is lacking [4]. Research indicates that three out of four women with POI retain ovarian follicles, albeit in a dormant state [5]. Consequently, there is an urgent need for novel drugs to awaken dormant primordial follicles. However, the unclear etiology of POI presents a barrier to drug development.

Although the exact mechanism underlying POI remains unknown, numerous studies have identified genetic factors as the most commonly implicated cause, offering potential targets for therapeutic interventions [6, 7]. Currently, the most commonly recognized genetic causes of POI include X chromosome abnormalities, notably Turner syndrome (13% of cases) [8], and FMR1 premutations (3–15% of cases) [9]. A clinical study involving 27 POI patients identified FMR1 premutations as a potential target for treating occult POI [10]. Similarly, a multicenter observational study involving 291 POI patients identified seven genes (USP36, VCP, WDR33, PIWIL3, NPM2, LLGL1, and BOD1L1) as potential targets for POI treatment [11]. However, it is essential to recognize that these results stem from observational studies, making them susceptible to confounding variables and reverse causation. Additionally, the constraints of observational studies limit the investigation of causal relationships between different genetic variants and POI, underscoring the necessity for more rigorous research approaches.

Genome-wide association analysis (GWAS) is pivotal for identifying single nucleotide polymorphisms (SNPs) and novel genetic variants associated with diseases. Nevertheless, the risk loci identified via GWAS are predominantly in non-coding regions of the genome, complicating their interpretation [12]. In contrast, expression quantitative trait loci (eQTL) studies quantify gene expression levels across the genome, offering an unbiased view of gene expression regulation [13]. Combining eQTL data with GWAS findings facilitates the identification of target genes driving the GWAS signal at specific loci [12]. In our study, we employed eQTL as an exposure tool in Mendelian randomization (MR) to examine causal links between genetic variants and POI. Moreover, we performed colocalization analyses to identify potential therapeutic targets for POI.

Methods

Study design

The study design is illustrated in Fig. 1. Cis-eQTL data were obtained from the Genotype-Tissue Expression (GTEx, https://gtexportal.org/home/) database[14] and the eQTLGen consortium (https://www.eqtlgen.org/)[15]. POI GWAS data were sourced from the FinnGen study (https://www.finngen.fi/en)[16]. As this research reanalyzed previously collected and published data, no further ethics approval was required.

Fig. 1
figure 1

Study design. PP.H3, the posterior probability of H3; PP.H4, the posterior probability of H4; HEIDI, instrument-dependent heterogeneity; MR, Mendelian Randomization

Acquisition of eQTL and GWAS Summary Data

Cis-eQTL data were utilized from GTEx V8 and the eQTLGen consortium [14, 15]. The GTEx V8 dataset included 838 primarily European participants and spanned 49 tissues or cell types. Specifically, cis-eQTL data from the ovary (n = 167) and whole blood (n = 670) were extracted within the GTEx V8 dataset. The eQTLGen consortium provided cis-eQTL data from peripheral blood samples of 31,684 participants. A PeQTL threshold of < 5 × 10− 8 was applied to all cis-eQTL. POI GWAS data were derived from the FinnGen study’s R11 dataset, comprising 599 cases and 241,998 controls [16]. All participants in this dataset were of European descent.

MR Analysis

We utilized the SMR software tool (version 1.3.1) to conduct SMR analysis on cis-eQTL and GWAS data, aiming to identify gene-POI associations [17]. To address potential pleiotropy between exposure and outcomes, we performed an instrument-dependent heterogeneity (HEIDI) test on the cis-eQTL and GWAS data. A PHEIDI < 0.05 indicates significant interlocking pleiotropy between two distinct genetic variants, leading to their exclusion from the study. Additionally, a two-sample MR analysis was conducted using gene-indexed SNPs, employing the Wald ratio and delta method to calculate odds ratios (OR) and 95% confidence intervals (CI) between genes and POI [17]. A Bonferroni-corrected P < 0.05 was used as the threshold to establish a causal relationship between genes and GWAS [18].

Colocalization analysis

The coloc R package was employed to perform colocalization analyses, assessing the impact of linkage disequilibrium on gene-POI associations [19]. Colocalization, using a Bayesian approach, produced posterior probabilities for five hypotheses: PP.H0 indicates no association with either trait; PP.H1 indicates an association with trait 1 only (gene expression); PP.H2 indicates an association with trait 2 only (POI trait); PP.H3 indicates an association with both traits but different causal variants for each; and PP.H4 indicates an association with both traits with the same causal variant [20]. We applied default priors: p1 = 1 × 10− 4, p2 = 1 × 10− 4, p12 = 1 × 10− 5. Low PP.H3 and PP.H4 combined with high PP.H0, PP.H1, and/or PP.H2 indicate limited power in the colocalization analysis. Therefore, we restricted our analysis to genes with PP.H3 + PP.H4 ≥ 0.8 [21].

Potential drug target identification

To evaluate the druggability of the genes, we queried Online Mendelian Inheritance in Man (OMIM), the DrugBank database, the Drug-Gene Interaction database (DGIdb), and the Therapeutic Target Database (TTD) to identify mutations linked to phenotypic abnormalities. Our evaluation criteria included: (1) approval for marketing or involvement in clinical trials, (2) preclinical development stage, and (3) considered druggable, even if not documented in the database but recognized as a potential drug target by our team [22].

Results

Genome-wide MR analysis

GWAS loci are often located in non-coding regions of the genome, making it difficult to interpret their functions [23]. Cis-eQTL analysis helps determine whether non-coding loci identified by GWAS affect gene expression, elucidating the relationship between gene expression and genetic variation and identifying potential candidate genes [24]. In this study, we explored the association between 431 genes with available index cis-eQTL signals and the risk of POI outcomes using a two-sample MR analysis (Supplementary Table S1). Associations with PHEIDI < 0.05 were deemed likely due to pleiotropy, resulting in the exclusion of 57 genes from the analysis. By applying a bonferroni-corrected P threshold of 0.05, we identified four genes with statistical significance. The MR analysis outcomes are illustrated in Fig. 2. Our findings predicted that HM13, FANCE, RAB2A, and MLLT10 were significantly associated with a reduced risk of POI (Table 1).

Fig. 2
figure 2

Manhattan plots for associations of genome-wide with POI in MR analysis. Labelled and red genes refer to MR findings with PHEIDI ≥ 0.05 and Bonferroni-corrected P < 0.05. (a) Associations of genetically predicted genes levels from Finngen_Whole_Blood_GTEx_V8 with POI; (b) Associations of genetically predicted genes levels from Finngen_Ovary_GTEx_V8 with POI; (c) Associations of genetically predicted genes levels from Finngen_eQTLGen with POI

Table 1 The mendelian randomization and colocalization results of cis-eQTL datasets and POI GWAS

Colocalization analysis

In MR studies, linkage disequilibrium (LD) can affect result accuracy. Colocalization analysis helps distinguish between true causal variants and those merely linked to causal variants, enhancing causal inference accuracy [19]. Evidence suggests that genes supported by both MR and colocalization evidence are more likely to be successful drug targets [25]. We also performed colocalization analyses to examine genes related to POI, investigating whether the genes identified in the FinnGen study shared genetic variants associated with POI. We found strong evidence of colocalization (PP.H3 + PP.H4 ≥ 0.8) for FANCE and RAB2A (Fig. 3) [21]. However, the posterior probability of HM13 and MLLT10 did not reach strong evidence of colocalization (PP.H3 + PP.H4 ≥ 0.8), likely due to insufficient samples in GTEx and eQTLGen.

Fig. 3
figure 3

High support evidence for colocalization between genes and POI. The size and colour of circle indicate the for PP.H3 + PP.H4

Druggability of identified genes

To determine the potential of genes identified by MR and colocalization analysis as drug targets, we specifically assessed FANCE and RAB2A, which showed strong evidence of colocalization. Although these genes have not been recognized as drug targets for POI in existing studies, they remain promising candidates (Supplementary Table S2). Mutations in FANCE and RAB2A are known to cause monogenic diseases. Specifically, mutations in the FANCE gene lead to Fanconi anemia (FA) [26], whereas no drug information was found for RAB2A. Our MR results suggest that inhibiting FANCE and RAB2A could potentially be a treatment for POI.

Discussion

Our study employed comprehensive genome-wide MR and colocalization analyses to clarify the causal relationships between genes and POI, providing valuable insights into potential therapeutic targets. MR analysis effectively reduced confounding factors in assessing the associations between gene expression and disease. Colocalization analysis confirmed that the eQTL instrument in the MR was not incidentally associated with both traits, thereby ruling out the possibility of the MR effect stemming from alternative causal variants in linkage disequilibrium. Among the genes analyzed, only FANCE and RAB2A exhibited evidence of a shared genetic effect with POI outcomes through both MR and colocalization analyses. FANCE and RAB2A were linked to a reduced risk of POI, highlighting their potential as promising targets for POI treatment.

FANCE is a subunit of the FA pathway, which plays a key role in repairing DNA interstrand cross-links. FA pathway genes encode proteins involved in gonadal development, DNA replication, and DNA repair [27]. Mutations in the FANCE gene cause FA in humans [28]. Our study confirmed that FANCE is associated with a reduced risk of POI and is a risk locus for POI. Interestingly, clinical evidence indicates that female FA patients exhibit reduced fertility, manifesting as POI [29]. Previous studies have shown that FANCE−/− mice exhibit ovarian dysplasia and severely reduced numbers of follicles by five days after birth, resembling women suffering from POI [30]. Animal experiments have also demonstrated that FANCE defects impair the rapid mitotic proliferation of primordial germ cells (PGCs) in mouse embryos, leading to a sharp decrease in PGCs number and abnormal cell cycle distribution [31]. These findings indicate that FANCE is essential for PGC survival, with potential mechanisms involving cell cycle regulation, DNA damage repair, cell death prevention, and the regulation of lysosome and ribosome functions [32].

In FA cells, DNA damage remains unrepaired due to a dysfunctional DNA repair process, causing cells to be blocked in the G2/M phase [33]. When DNA damage occurs in the primordial follicle, it responds by phosphorylating and activating TAp63 [34]. Activation of TAp63 induces the transcription of pro-apoptotic factors such as BH3, PUMA and NOXA. The upregulated expression of these proteins facilitates their interaction with the pro-apoptotic BCL2 family members, BAX and BAK [34]. The translocation of BAX and/or BAK to the oocyte’s mitochondria causes mitochondrial dysfunction, release of apoptogenic proteins, and activation of caspase-9 and proteolytic enzymes, collectively triggering apoptosis and cell death [34]. The depletion of oocytes damages fertility and leads to POI [35]. Two rounds of meiosis are vital for oocyte maturation [36]. During the prophase of the first meiosis, fully grown oocytes are arrested in the germinal vesicle (G2) phase [37]. Meiotic resumption marks the initiation of oocyte maturation, characterized by germinal vesicle breakdown (GVBD), followed by meiotic spindle assembly and migration during metaphase I (MI)[38]. Subsequently, cytokinesis occurs, the oocyte extrudes the first polar body, and arrests in the metaphase II (MII) phase [37]. The G2/M phase and the MII phase are two major stages in oocyte meiosis [38]. G2/M cell cycle blockade leads to meiotic recovery failure, chromosome misalignment, increased aneuploidy, abnormal spindle assembly, and severe meiotic defects in oocytes [37, 39, 40]. These issues are the main causes of ovarian aging and reduced female fertility [41].

RAB2A, a member of the Rab family, is localized to the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) transport complex and regulates ERGIC transport within the cell membrane [42]. Our findings indicate that genetically predicted RAB2A is inversely associated with the risk of POI. The HEIDI test and colocalization analysis further ruled out the possibility of horizontal pleiotropy. While current studies show that RAB2A is associated with sperm viability and motility function in men [43], the mechanism of premature ovarian aging in females with POI remains unclear.

Numerous studies have shown that ovarian aging is associated with mitochondria, oxidative stress, DNA damage, protein homeostasis, aneuploidy, apoptosis, and autophagy [44]. Previous research has demonstrated that RAB2A regulates autophagosome-lysosome fusion [45]. RAB2 may regulate autophagy initiation through three mechanisms: (1) Transporting Golgi-derived ATG9 + vesicles to phagophore assembly sites. (2) Recruiting ULK1 to phagophore assembly sites, as ULK1 appears soluble and forms a diffused cytosolic pattern in the absence of RAB2A. (3) Facilitating ULK1 activation to propagate signals for autophagy initiation [45]. Autophagy plays a crucial role in oocyte development. Under normal circumstances, oocytes cannot actively induce mitophagy to clear damaged mitochondria. However, oocyte mitophagy can be initiated by drugs or abnormal environmental stimuli, affecting the developmental ability of oocytes [46]. Based on our findings and literature reports, we hypothesize that high expression of RAB2A may induce abnormal autophagy in oocyte mitochondria, resulting in a decrease in the number and quality of oocytes, ultimately leading to POI. However, this hypothesis requires further study as no correlation has been reported in the literature yet.

The strength of our study lies in employing MR and colocalization analyses to evaluate gene causality in POI records through genetic variation. MR analysis reduces biases from confounders and reverse causation, thereby increasing the reliability of causality results. Colocalization analysis is instrumental in revealing the pleiotropic effects of specific loci on multiple traits, avoiding LD and identifying potential therapeutic targets. Additionally, we utilized extensive GWAS data, allowing the simultaneous examination of numerous genes and variants. This cost-effective and efficient method thoroughly investigates the gene-disease relationship. Our study integrated multiple databases, enhancing the robustness of our findings. Focusing on European populations minimized bias related to ethnic disparities.

Nonetheless, certain limitations should be acknowledged. Firstly, our colocalization analyses are constrained by the availability of outcomes from existing studies, limiting the examination of undiscovered genetic variants. Moreover, the reliance on instrumental variants in colocalization analysis can introduce bias if these variants correlate with unmeasured variables. Finally, while focusing on specific ethnicities reduces racial bias, it also limits the generalizability of our findings to diverse ethnic groups.

Conclusion

Our study has successfully established a causal relationship between genes and POI through comprehensive genome-wide MR and colocalization analyses. We identified FANCE and RAB2A as potential drug targets for POI treatment. However, these findings require validation in future trials.

Data availability

All data analysed in this study can be obtained by a reasonable request to corresponding authors.

Abbreviations

POI:

Primary ovarian insufficiency

HRT:

Hormone replacement therapy

GWAS:

Genome-wide association analysis

SNP:

Single nucleotide polymorphisms

eQTL:

expression quantitative trait loci

MR:

Mendelian randomization

HEIDI:

Instrument-dependent heterogeneity

CI:

Confidence intervals

OR:

Odds ratios

OMIM:

Online Mendelian Inheritance in Man

DGIdb:

The Drug-Gene Interaction database

TTD:

Therapeutic Target Database

HM13:

Histocompatibility minor 13

FANCE:

FA complementation group E

RAB2A:

RAB2A, member RAS oncogene family

MLLT10:

MLLT10 histone lysine methyltransferase DOT1L cofactor

LD:

Linkage disequilibrium

FA:

Fanconi anemia

PGCs:

Primordial germ cells

BH3:

BCL2 homology 3

PUMA:

p53-upregulated modulator of apoptosis

G2:

Germinal vesicle

GVBD:

Germinal vesicle breakdown

MI:

Metaphase I

MII:

Metaphase II

ERGIC:

Endoplasmic reticulum-Golgi intermediate compartment

References

  1. Stuenkel CA, Gompel A. Primary ovarian insufficiency. N Engl J Med. 2023;388(2):154–63. https://doi.org/10.1056/NEJMcp2116488

    Article  PubMed  Google Scholar 

  2. Faubion SS, Kuhle CL, Shuster LT, Rocca WA. Long-term health consequences of premature or early menopause and considerations for management. Climacteric. 2015;18(4):483–91. https://doi.org/10.3109/13697137.2015.1020484

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Golezar S, Ramezani Tehrani F, Khazaei S, Ebadi A, Keshavarz Z. The global prevalence of primary ovarian insufficiency and early menopause: a meta-analysis. Climacteric. 2019;22(4):403–11. https://doi.org/10.1080/13697137.2019.1574738

    Article  CAS  PubMed  Google Scholar 

  4. Committee Opinion No. 698: hormone therapy in primary ovarian insufficiency. Obstet Gynecol. 2017;129(5):e134–41. https://doi.org/10.1097/AOG.0000000000002044

    Article  Google Scholar 

  5. De Vos M, Devroey P, Fauser BC. Primary ovarian insufficiency. Lancet. 2010;376(9744):911–21. https://doi.org/10.1016/S0140-6736(10)60355-8

    Article  PubMed  Google Scholar 

  6. Fortuño C, Labarta E. Genetics of primary ovarian insufficiency: a review. J Assist Reprod Genet. 2014;31(12):1573–85. https://doi.org/10.1007/s10815-014-0342-9

    Article  PubMed  PubMed Central  Google Scholar 

  7. Huhtaniemi I, Hovatta O, La Marca A, et al. Advances in the Molecular Pathophysiology, Genetics, and treatment of primary ovarian insufficiency. Trends Endocrinol Metab. 2018;29(6):400–19. https://doi.org/10.1016/j.tem.2018.03.010

    Article  CAS  PubMed  Google Scholar 

  8. Cordts EB, Christofolini DM, Dos Santos AA, Bianco B, Barbosa CP. Genetic aspects of premature ovarian failure: a literature review. Arch Gynecol Obstet. 2011;283(3):635–43. https://doi.org/10.1007/s00404-010-1815-4

    Article  PubMed  Google Scholar 

  9. Wittenberger MD, Hagerman RJ, Sherman SL, et al. The FMR1 premutation and reproduction. Fertil Steril. 2007;87(3):456–65. https://doi.org/10.1016/j.fertnstert.2006.09.004

    Article  CAS  PubMed  Google Scholar 

  10. Streuli I, Fraisse T, Ibecheole V, Moix I, Morris MA, de Ziegler D. Intermediate and premutation FMR1 alleles in women with occult primary ovarian insufficiency. Fertil Steril. 2009;92(2):464–70. https://doi.org/10.1016/j.fertnstert.2008.07.007

    Article  CAS  PubMed  Google Scholar 

  11. Gorsi B, Hernandez E, Moore MB, et al. Causal and Candidate Gene Variants in a large cohort of women with primary ovarian insufficiency. J Clin Endocrinol Metab. 2022;107(3):685–714. https://doi.org/10.1210/clinem/dgab775

    Article  PubMed  Google Scholar 

  12. Schwarz T, Boltz T, Hou K, et al. Powerful eQTL mapping through low-coverage RNA sequencing. HGG Adv. 2022;3(3):100103. https://doi.org/10.1016/j.xhgg.2022.100103

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhang J, Zhao H. eQTL studies: from bulk tissues to single cells. J Genet Genomics. 2023;50(12):925–33. https://doi.org/10.1016/j.jgg.2023.05.003

    Article  PubMed  Google Scholar 

  14. GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science. 2020;369(6509):1318–30. https://doi.org/10.1126/science.aaz1776

    Article  CAS  Google Scholar 

  15. Võsa U, Claringbould A, Westra HJ, et al. Large-scale cis- and trans-eQTL analyses identify thousands of genetic loci and polygenic scores that regulate blood gene expression. Nat Genet. 2021;53(9):1300–10. https://doi.org/10.1038/s41588-021-00913-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Kurki MI, Karjalainen J, Palta P, et al. FinnGen provides genetic insights from a well-phenotyped isolated population. Nature. 2023;613:508–18. https://doi.org/10.1038/s41586-022-05473-8

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Zhu Z, Zhang F, Hu H, et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat Genet. 2016;48(5):481–7. https://doi.org/10.1038/ng.3538

    Article  CAS  PubMed  Google Scholar 

  18. Chen J, Xu F, Ruan X, et al. Therapeutic targets for inflammatory bowel disease: proteome-wide mendelian randomization and colocalization analyses. EBioMedicine. 2023;89:104494. https://doi.org/10.1016/j.ebiom.2023.104494

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Giambartolomei C, Vukcevic D, Schadt EE et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 2014;10(5):e1004383. Published 2014 May 15. https://doi.org/10.1371/journal.pgen.1004383

  20. Foley CN, Staley JR, Breen PG, et al. A fast and efficient colocalization algorithm for identifying shared genetic risk factors across multiple traits. Nat Commun. 2021;12(1):764. https://doi.org/10.1038/s41467-020-20885-8. Published 2021 Feb 3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Storm CS, Kia DA, Almramhi MM, et al. Finding genetically-supported drug targets for Parkinson’s disease using mendelian randomization of the druggable genome. Nat Commun. 2021;12(1):7342. https://doi.org/10.1038/s41467-021-26280-1. Published 2021 Dec 20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Finan C, Gaulton A, Kruger FA, et al. The druggable genome and support for target identification and validation in drug development. Sci Transl Med. 2017;9(383):eaag1166. https://doi.org/10.1126/scitranslmed.aag1166

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Montgomery SB, Sammeth M, Gutierrez-Arcelus M, et al. Transcriptome genetics using second generation sequencing in a caucasian population. Nature. 2010;464(7289):773–7. https://doi.org/10.1038/nature08903

    Article  CAS  PubMed  Google Scholar 

  24. Mancuso N, Shi H, Goddard P, Kichaev G, Gusev A, Pasaniuc B. Integrating Gene expression with Summary Association Statistics To Identify Genes Associated with 30 complex traits. Am J Hum Genet. 2017;100(3):473–87. https://doi.org/10.1016/j.ajhg.2017.01.031

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhao G, Lu Z, Liao Y, et al. Association of intestinal anti-inflammatory drug target genes with psychiatric disorders: a mendelian randomization study. J Adv Res Published Online May. 2024;11. https://doi.org/10.1016/j.jare.2024.05.002

  26. de Winter JP, Léveillé F, van Berkel CG, et al. Isolation of a cDNA representing the fanconi anemia complementation group E gene. Am J Hum Genet. 2000;67(5):1306–8. https://doi.org/10.1016/S0002-9297(07)62959-0

    Article  PubMed  PubMed Central  Google Scholar 

  27. Tsui V, Crismani W. The Fanconi Anemia Pathway and Fertility. Trends Genet. 2019;35(3):199–214. https://doi.org/10.1016/j.tig.2018.12.007

    Article  CAS  PubMed  Google Scholar 

  28. Zheng C, Ren Z, Chen H, et al. Reduced FANCE confers genomic instability and malignant behavior by regulating cell cycle progression in Endometrial Cancer. J Cancer. 2023;14(14):2670–85. https://doi.org/10.7150/jca.86348

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Auerbach AD. Fanconi anemia and its diagnosis. Mutat Res. 2009;668(1–2):4–10. https://doi.org/10.1016/j.mrfmmm.2009.01.013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Fu C, Begum K, Overbeek PA. Primary ovarian Insufficiency Induced by Fanconi Anemia E mutation in a mouse model. PLoS ONE. 2016;11(3):e0144285. https://doi.org/10.1371/journal.pone.0144285. Published 2016 Mar 3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhou Z, Yin H, Suye S, et al. Fance deficiency inhibits primordial germ cell proliferation associated with transcription-replication conflicts accumulate and DNA repair defects. J Ovarian Res. 2023;16(1):160. https://doi.org/10.1186/s13048-023-01252-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Suye S, Yin H, Zhou Z, et al. Histological and transcriptomic analysis of Fance-deficient PGCs reveal the possible mechanisms of their depletion. Reproduction. 2023;166(1):65–75. https://doi.org/10.1530/REP-23-0038. Published 2023 Jun 12.

    Article  CAS  PubMed  Google Scholar 

  33. Sala-Trepat M, Rouillard D, Escarceller M, Laquerbe A, Moustacchi E, Papadopoulo D. Arrest of S-phase progression is impaired in fanconi anemia cells. Exp Cell Res. 2000;260(2):208–15. https://doi.org/10.1006/excr.2000.4994

    Article  CAS  PubMed  Google Scholar 

  34. Kerr JB, Hutt KJ, Michalak EM, et al. DNA damage-induced primordial follicle oocyte apoptosis and loss of fertility require TAp63-mediated induction of Puma and Noxa. Mol Cell. 2012;48(3):343–52. https://doi.org/10.1016/j.molcel.2012.08.017

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Martin JH, Aitken RJ, Bromfield EG, Nixon B. DNA damage and repair in the female germline: contributions to ART. Hum Reprod Update. 2019;25(2):180–201. https://doi.org/10.1093/humupd/dmy040

    Article  CAS  PubMed  Google Scholar 

  36. MacLennan M, Crichton JH, Playfoot CJ, Adams IR. Oocyte development, meiosis and aneuploidy. Semin Cell Dev Biol. 2015;45:68–76. https://doi.org/10.1016/j.semcdb.2015.10.005

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang KH, Zou YJ, Shan MM, et al. Arf1 GTPase regulates golgi-dependent G2/M transition and Spindle Organization in Oocyte Meiosis. Adv Sci (Weinh). 2024;11(4):e2303009. https://doi.org/10.1002/advs.202303009

    Article  CAS  PubMed  Google Scholar 

  38. Duan X, Sun SC. Actin cytoskeleton dynamics in mammalian oocyte meiosis. Biol Reprod. 2019;100(1):15–24. https://doi.org/10.1093/biolre/ioy163

    Article  PubMed  Google Scholar 

  39. Koliopoulos MG, Alfieri C. Cell cycle regulation by complex nanomachines. FEBS J. 2022;289(17):5100–20. https://doi.org/10.1111/febs.16082

    Article  CAS  PubMed  Google Scholar 

  40. Zhao BW, Sun SM, Xu K, et al. FBXO34 regulates the G2/M transition and anaphase entry in meiotic oocytes. Front Cell Dev Biol. 2021;9:647103. https://doi.org/10.3389/fcell.2021.647103. Published 2021 Mar 25.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Wasielak-Politowska M, Kordowitzki P. Chromosome segregation in the oocyte: what goes wrong during aging. Int J Mol Sci. 2022;23(5):2880. https://doi.org/10.3390/ijms23052880. Published 2022 Mar 7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sugawara T, Kano F, Murata M. Rab2A is a pivotal switch protein that promotes either secretion or ER-associated degradation of (pro)insulin in insulin-secreting cells. Sci Rep. 2014;4:6952. https://doi.org/10.1038/srep06952. Published 2014 Nov 7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Bae JW, Hwang JM, Kwon WS. Prediction of male fertility using ras-related proteins. J Anim Sci Technol. 2022;64(6):1024–34. https://doi.org/10.5187/jast.2022.e83

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Wang X, Wang L, Xiang W. Mechanisms of ovarian aging in women: a review. J Ovarian Res. 2023;16(1):67. https://doi.org/10.1186/s13048-023-01151-z. Published 2023 Apr 6.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Ding X, Jiang X, Tian R, et al. RAB2 regulates the formation of autophagosome and autolysosome in mammalian cells. Autophagy. 2019;15(10):1774–86. https://doi.org/10.1080/15548627.2019.1596478

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shen Q, Liu Y, Li H, Zhang L. Effect of mitophagy in oocytes and granulosa cells on oocyte quality†. Biol Reprod. 2021;104(2):294–304. https://doi.org/10.1093/biolre/ioaa194

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to express our sincere gratitude to the dedicated participants and meticulous investigators who contributed to the Genotype-Tissue Expression project, the eQTLGen Consortium, and the FinnGen study.

Funding

This work was supported by the Scientific Research Project of Sichuan Maternal and Child Health Association (no.22FXYB12; no.22FXYB02).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, HD, PZ and ZH; Formal analysis, HD; Methodology, HD and XL; Resources, ZH and JZ; Supervision, JZ; Visualization, HD and PZ; Writing-original draft, HD; Writing-review & editing, PZ, XL, JZ, and ZH. All authors reviewed the manuscript.

Corresponding author

Correspondence to Zhonglu Huang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, H., Zeng, P., Liu, X. et al. Identifying therapeutic targets for primary ovarian insufficiency through integrated genomic analyses. J Ovarian Res 17, 193 (2024). https://doi.org/10.1186/s13048-024-01524-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13048-024-01524-y

Keywords