Next Article in Journal
WHITE MATTER MATTERS: New Approach to the Brain’s Hidden Half Using Circulating Oligodendrocyte-Derived Extracellular Vesicles
Previous Article in Journal
Functional and In Silico Characterization of ALPL Gene Variants Reveals Genotype–Phenotype Correlations in Italian Hypophosphatasia Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CRISPR as a Tool to Uncover Gene Function in Polycystic Ovary Syndrome: A Literature Review of Experimental Models Targeting Ovarian and Metabolic Genes

1
School of Medicine, Royal College of Surgeons in Ireland, Medical University of Bahrain, Busaiteen P.O. Box 15503, Bahrain
2
Research Department, Royal College of Surgeons in Ireland, Medical University of Bahrain, Busaiteen P.O. Box 15503, Bahrain
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2025, 14(22), 1769; https://doi.org/10.3390/cells14221769
Submission received: 30 September 2025 / Revised: 26 October 2025 / Accepted: 29 October 2025 / Published: 12 November 2025
(This article belongs to the Special Issue AI, CRISPR, and Molecular Mechanisms in Precision Medicine)

Highlights

What are the main findings?
  • CRISPR models reveal gene-specific contributions to ovarian and metabolic PCOS phenotypes, including hyperandrogenism and insulin resistance.
  • Single-gene models show species-specific effects, highlighting the need for dual-gene and integrated models.
What are the implications of the main findings?
  • CRISPR studies identify potential diagnostic markers and clarify gene-hormone interactions in PCOS.
  • Combined gene editing and hormonal models provide a platform for studying the polygenic, multifactorial nature of PCOS.

Abstract

Polycystic ovary syndrome (PCOS) is a complex disorder characterized by reproductive abnormalities such as hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology, and is frequently accompanied by metabolic disturbances such as insulin resistance, obesity and dyslipidemia. Genome-wide association studies (GWASs) have identified several susceptibility loci, yet little is known about their functional implications. Clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9) has emerged as a powerful gene editing tool in bridging this gap by allowing researchers to directly target candidate genes in ovarian and metabolic pathways. For instance, experimental models have highlighted the role of CYP17A1 and DENND1A.V2 in androgen excess, anti-Müllerian hormone (AMH) in follicular arrest, and insulin receptor substrate 1 (IRS1) and PPARγ in insulin signaling and adipogenesis. To highlight the multifactorial nature of PCOS, animal models, including zebrafish and rodents, have been used to reveal interactions between reproductive and metabolic phenotypes. Nevertheless, most studies remain restricted to single-gene models, and dual-gene models or combined gene editing and hormonal induction models remain underexplored. Future research integrating precision editing, multi-omic platforms, and patient-derived organoids may provide more accurate disease models and novel therapeutic strategies.

1. Introduction

Polycystic ovary syndrome (PCOS) is a complex heterogeneous disorder characterized by clinical and biochemical features of androgen excess and disturbances in ovulatory function, primarily affecting females aged 15 to 49 years [1]. Its global prevalence among premenopausal women is estimated at 6–20%, or approximately 1 in 15 females [1]. PCOS represents a leading cause of female infertility and is commonly associated with clinical manifestations such as menstrual irregularities, hirsutism, cystic acne, seborrhea, alopecia and obesity [2]. Beyond reproductive features, the syndrome is often accompanied by metabolic dysfunctions, including insulin resistance, obesity, dyslipidemia, type 2 diabetes mellitus (T2DM) and heightened cardiovascular risk [3]. PCOS has emerged as the primary contributor to anovulatory infertility and, given its high prevalence, imposes significant health and economic burdens on both families and society [3].
From a diagnostic perspective, the Rotterdam criteria remain the principal diagnostic standard, requiring at least two of the following features are present: oligo-ovulation or anovulation, clinical or biochemical hyperandrogenism and polycystic ovaries [1]. Current guidelines recommend characterizing PCOS according to four phenotypes (A–D), which are defined by the presence or absence of the three diagnostic features: hyperandrogenism, ovulatory dysfunction, and polycystic ovarian morphology (PCOM) [4]. Moreover, the phenotypes of PCOS are classified into: phenotype A, meeting all three diagnostic criteria; phenotype B, defined by anovulation and hyperandrogenism; phenotype C, comprising hyperandrogenism with PCOM; and phenotype D, characterized by anovulation with PCOM [5].
PCOS is a complex, multifactorial endocrinopathy that reflects an interplay between genetic predisposition, ovarian dysfunction, neuroendocrine dysregulation, metabolic disturbances and chronic low-grade inflammation [6]. At the endocrine level, PCOS leads to an increased gonadotropin-releasing hormone (GnRH) pulse frequency that preferentially stimulates luteinizing hormone (LH) secretion follicle-stimulating hormone (FSH) secretion [7]—reported in up to 94% of patients [8]. Persistently elevated LH overstimulates ovarian theca cells, upregulating CYP17A1, CYP11A1 and HSD3B2, which enhance androgen synthesis [9,10,11]. Meanwhile, low FSH impairs aromatase (CYP19A1) activity, reducing androgen-to-estrogen conversion and causing follicular arrest [12]. Accumulation of these arrested follicles produces the characteristic polycystic ovarian morphology [1]. Hyperandrogenism, present in over 75% of PCOS cases, leads to follicular atresia, impairs the proliferation of cumulus cells, disrupts angiogenesis, and compromises oocyte maturation [13,14,15,16,17].
Insulin resistance (IR) affects 50–70% of women with PCOS, regardless of body mass index (BMI), and is often accompanied by compensatory hyperinsulinemia [18,19]. Hyperins
Hyperinsulinemia synergizes with LH on theca cells, upregulating CYP17A1 transcription and cytochrome P450 17α-hydroxylase (P450c17α) activity while simultaneously lowering hepatic sex hormone-binding globulin (SHBG) and insulin-like growth factor-binding protein-1 (IGFBP-1) [11,20,21,22,23,24,25,26]. Systematically, IR contributes to the threefold higher prevalence of metabolic syndrome in PCOS, increasing the risk of hypertension, dyslipidemia, obesity and hyperglycemia [27]. Importantly, lifestyle changes or insulin-sensitizing agents such as metformin decrease androgen levels and improve ovulatory function [28,29,30].
Chronic low-grade inflammation is a central feature of PCOS, acting alongside hyperandrogenism and IR [31]. Women with PCOS demonstrate increased C-reactive protein (CRP), interleukin-6 (IL-6), interleukin-18 (IL-18), tumor necrosis factor-α (TNF-α) and white blood cell counts together with dysregulation of pro- and anti-inflammatory mediators in serum and follicular fluid [31,32,33,34,35,36,37]. Elevated cytokines are seen even in lean patients, suggesting inflammation as an intrinsic rather than an obesity-driven factor [38,39,40,41]. Excessive ovarian inflammation reduces oocyte quality, impairs folliculogenesis, and contributes to anovulation [42].
Hyperandrogenism further induces inflammation in visceral adipose tissue, a key source of IL-6, TNF-α, and leptin, which promote low-grade chronic inflammation, impair insulin signaling, and exacerbate IR [11,43]. Hyperandrogenism also activates nuclear factor kappa B (NF-κB), amplifying inflammatory pathways [44]. Epigenetic regulators, particularly exosomal microRNAs (miRNAs) (e.g., miR-155-5p, miR-222), modulate the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt)-GLUT4 pathway and correlate with insulin levels, indicating biomarker potential [45]. This inflammatory state underpins both metabolic complications and increased cardiovascular risk, emphasizing the importance of integrating anti-inflammatory strategies into PCOS management [46,47]. The key reproductive, metabolic, and inflammatory mechanisms underlying PCOS are summarized in Figure 1.
Recent clinical management of PCOS centers on a combination of lifestyle modification and pharmacotherapy to target reproductive and metabolic features [48]. Pharmacological options include combined oral contraceptives and anti-androgens (e.g., spironolactone) to manage hyperandrogenic symptoms, and insulin-sensitizing agents such as metformin which improve insulin resistance and can restore ovulation in some women [48]. For infertility due to anovulation, aromatase inhibition with letrozole has surpassed clomiphene as the preferred first-line ovulation induction agent in many guidelines [49]. Emerging and increasingly studied therapies include incretin-based GLP-1 receptor agonists (e.g., liraglutide, semaglutide) and SGLT2 inhibitors, which have shown promising effects on weight, insulin resistance and some endocrine parameters in short-term trials [50]; inositol is also supported by systematic reviews as a modestly beneficial adjunct for metabolic and ovulatory outcomes in selected patients [51]. While these approaches do not yet constitute disease-modifying therapies for the underlying polygenic and endocrine drivers of PCOS, they provide clinically meaningful improvements in reproductive and metabolic outcomes and are active areas of translational research.
Furthermore, gene editing technology such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (CRISPR/Cas9) will be explored to investigate its potential therapeutic applications. CRISPR/Cas is identified as an adaptive immune mechanism in bacteria and archaeans, which is now being used as gene editing technology [52]. This review aims to provide a comprehensive overview of PCOS, with particular emphasis on the potential use of CRISPR technology in advancing PCOS research.

Genetic Landscape of PCOS

Over the last decade, genome-wide association studies (GWASs) have significantly advanced our understanding of the genetic underpinnings of PCOS [53]. Studies have identified over 20 susceptibility loci for PCOS across populations, most prominently in DENND1A, THADA, LHCGR, FSHR, INSR, YAP1, RAB5B and HMGA2 [53,54]. These genes converge on pathways critical to gonadotropin signaling, ovarian steroidogenesis, insulin signaling and metabolic regulation [55,56]. DENND1A, in particular, has emerged as a pivotal candidate, with the splice isoform DENND1A.V2 functionally linked to excess androgen production in PCOS theca cells [57,58]. Similarly, THADA variants have been associated with impaired energy metabolism, while INSR variants implicate abnormal insulin signaling, underscoring the metabolic dimension of PCOS [59,60]. Furthermore, FSHR and LHCGR polymorphisms highlight dysregulated folliculogenesis and gonadotropin response [53,61,62,63].
Recent studies have expanded the genetic landscape to include KISS1, LEPR, SOD2, ERBB4, WWTR1, CHEK2 and ADIPOQ, broadening the understanding of PCOS [53]. KISS1, which encodes kisspeptin, regulates GnRH secretion and follicular maturation [64,65]. Polymorphisms such as rs4889 have been associated with altered BMI, lower LH and GnRH feedback disruption, with population-level associations in Saudi Arabian women [64,65]. This has functional implications, which include altered kisspeptin neuron morphology in PCOS models and therapeutic exploration of kisspeptin receptor agonists to induce ovulation [66,67,68]. Variants in LEPR (rs1137100, rs1137101) affect the binding of leptin and downstream pathways of energy balance and insulin sensitivity [69,70]. Notably, rs1137101 has been shown to have a protective metabolic profile, whereas rs1137100 correlates with increased IR, particularly shown in Bahraini cohorts [69,71]. In addition, SOD2, through its A16V variant, has been linked to oxidative stress in Chinese women with PCOS, potentially contributing to cellular injury and abnormal gonadotropin ratios [71,72,73,74]. In European and Han Chinese populations, ERBB4 polymorphisms (e.g., rs113168128) have been reported to be consistently associated with these populations [75,76]. ERBB4 knockout models in granulosa cells replicate key features of PCOS, such as hyperandrogenism, menstrual cycle disturbances, and metabolic alterations [77]. The Hippo pathway component WWTR1, particularly the variant rs144248326, has been associated with infertility and oligomenorrhea [72,78]. Along with YAP1, WWTR1 also regulates IR, and its inhibition enhances the efficacy of metformin [79]. Moreover, CHEK2 gene variants are involved in regulating ovarian reserve, where rare frameshift/missense mutations may disrupt oocyte apoptosis, delaying reproductive senescence and mirroring the prolonged ovarian reserve seen in PCOS [80,81,82,83]. Modulating adiponectin signaling also contributes to PCOS susceptibility [84]. This has been linked to adiponectin (ADIPOQ) and adiponectin receptor 1 (ADIPOR1) genes [84,85]. Single nucleotide polymorphisms (SNPs) in ADIPOQ show population-specific associations in which six specific SNPs have been reported to have a significant association with the development of PCOS in Bahraini women [86]. However, results seem to differ by ethnicity when looking at ADIPOQ gene variants. Specifically, studies in Saudi Arabian and Jordanian women did not show the same association between ADIPOQ variants and PCOS as was seen in Bahraini women [87,88]. Additionally, polymorphisms in ADIPOR1 have been linked to IR in Chinese cohorts [85]. Importantly, in PCOS mouse models, adiponectin treatment ameliorates hyperandrogenism and metabolic dysfunction [89].
Although GWAS has progressed, findings face inherent limitations. Gene prioritization is complicated, as most signals map to non-coding regions [90,91]. Moreover, linkage disequilibrium obscures causality, and effect sizes remain small, accounting for only part of PCOS heritability [55]. In addition, ethnic diversity remains underrepresented, with most data derived from East Asian and European populations [92,93,94]. Furthermore, studies fail to distinguish between the clinical subtypes of PCOS, which may confound results and obscure subtype-specific genetic associations [95]. Finally, the phenotypic heterogeneity in PCOS limits reproducibility between different study cohorts [56]. Functional validation is essential to bridge the gap between association and mechanism. The DENND1A.V2 model demonstrates this, as it shows a direct causal effect between a GWAS locus and hyperandrogenism [57]. Genome-editing tools such as CRISPR/Cas9 offer powerful strategies to model genetic variants within endocrine and metabolic systems, thereby clarifying their roles in cell signaling and endocrine regulation [96]. Recent advances in genetic and molecular studies are summarized in Table 1, which highlights key genes implicated in PCOS pathophysiology and their potential relevance to CRISPR-based applications.

2. Overview of CRISPR Technology and Its Applications in Endocrine Research

CRISPR-based technologies are key tools in endocrine and reproductive research, where precise gene regulation is crucial. The CRISPR/Cas system was adapted from bacterial and archaeal immune defense mechanisms, where it functioned to recognize and cleave the DNA of invading viruses [99]. It was then redesigned as a tool that enabled researchers to modify genetic material in various organisms [100]. Among the various types identified, CRISPR/Cas9 is a type II system that requires a single Cas9 guided by single guide RNA (sgRNA) [101]. The sgRNA directs Cas9 to a genomic target adjacent to a protospacer adjacent motif (PAM) sequence, where the nuclease induces a double-stranded break in the target DNA [100,102,103].
Before the advent of CRISPR/Cas systems, several techniques were used for the manipulation of genetic material. Among these was homologous recombination (HR), a natural process whereby genetic material is exchanged between genomic and exogenous DNA through the crossing over of homologous DNA strands [104]. HR allows for modifications to be introduced to specific genes; however, it is considered to be inaccurate, inefficient and time-consuming [104]. Over time, technological advances led to the development of engineered nucleases, such as zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), which overcame HR [105]. However, ZFNs and TALENs also proved to be problematic due to their complexity, tedious design, high cost and labor-intensive nature [99]. Therefore, the techniques are not suitable for large-scale studies [99]. On the other hand, the CRISPR/Cas system only requires the guide RNA (gRNA) to be modified and constructed, which is faster and easier to design than proteins [99]. In addition, its capacity for multiplexed genome editing, where multiple loci are targeted simultaneously, makes it a more efficient, practical and scalable tool for biomedical research compared to ZFNs and TALENs [106].
These features have made CRISPR particularly valuable, pushing forward reproductive biology by facilitating functional genomics and mutation repair in animal models, and showing promise in the treatment of infertility and genetic diseases [107]. In recent studies, targeted gene editing has allowed researchers to manipulate the activity of hormone receptors, enabling them to understand how these changes affect ovarian function. For instance, Guller et al. applied a CRISPR/dCas9 approach to increase FSHR in human granulosa cells, making them more responsive to FSH and activating important signaling pathways [108]. Most importantly, the study highlights the potential of CRISPR to model and investigate FSH-related conditions, such as PCOS, in vitro [108].
Furthermore, CRISPR has been instrumental in metabolic research by allowing for the precise editing of genes that regulate insulin signaling and glucose metabolism. An estimated 70% of women with PCOS experience IR accompanied by hyperinsulinemia, which increases their risk of developing T2DM and makes these abnormalities a prime candidate for gene-based therapies [109]. In T2DM, IR is characterized by polymorphisms in genes that control insulin signaling pathways, such as IRS1 and IRS2 [110]. Through CRISPR, the function of IRS1 and IRS2 can be enhanced to increase insulin sensitivity in skeletal muscle and adipose tissue, thus improving glucose uptake [110]. Moreover, Matboli et al. have identified a competing endogenous RNA (ceRNA) network of long non-coding RNAs (lncRNAs), miRNAs, and mRNAs linked to IR, along with their target signaling pathways genes [111]. In T2DM, this ceRNA network becomes dysregulated and contributes to impaired insulin signaling [111]. Using CRISPR/Cas9, the researchers knocked out one of the lncRNAs and restored normal expression of the genes linked to T2DM, highlighting a novel approach to the treatment of the disease [111].
These applications demonstrate the versatility of CRISPR as a gene editing tool and its potential for use in conditions such as PCOS, which lies at the intersection between ovarian dysfunction and metabolic disturbance.

3. CRISPR in PCOS-Related Ovarian Gene Function

3.1. Theca Cell-Specific Genes

Theca cells form the outer layer of developing ovarian follicles and are specialized for androgen biosynthesis [112]. Under LH stimulation, they express steroidogenic enzymes such as CYP17A1 and CYP11A1, which convert cholesterol into androgen precursors later aromatized into estrogens by granulosa cells [113]. In PCOS, theca cell hyperresponsiveness and dysregulated steroidogenesis contribute directly to systemic hyperandrogenism [114,115]. Genetic studies have implicated theca-enriched genes in this phenotype, but functional validation has been limited. CRISPR-based models now offer tools to interrogate these candidate genes with cell-type precision [116].
DENND1A.V2, an isoform of the GWAS-nominated DENND1A gene, is consistently upregulated in PCOS theca cells and confers a pro-androgenic transcriptomic profile [117]. Overexpression of DENND1A.V2 in human theca-like cells enhanced CYP17A1 expression and androgen output, mirroring PCOS-associated hyperandrogenism [57]. While global knockout of DENND1A results in embryonic lethality, the potential for CRISPR-based conditional models or humanized knock-ins may allow interrogation of DENND1A.V2 in vivo [118].
A more direct CRISPR model was developed for CYP17A1, a key enzyme in androgen biosynthesis. Using a doxycycline-inducible CRISPR-Tet-On system, Joseph et al. overexpressed CYP17A1 specifically in mouse ovarian theca cells [119]. The resultant TC17 females developed marked hyperandrogenism, prolonged estrous cycles and cystic follicle morphology, features that mirror clinical and histologic findings in PCOS [119]. Notably, these defects were reversible upon doxycycline withdrawal, validating this as a temporally controlled, high-fidelity model of androgen-driven PCOS [119].
Furthermore, emerging targets involved in cholesterol trafficking and adrenocortical tropic hormone (ACTH) sensitivity, such as melanocortin 2 receptor (MC2R), steroidogenic acute regulatory protein (StAR) and CYP11A1, also exhibit differential expression in theca cells [120,121,122].

3.2. Granulosa Cell-Related Genes

Granulosa cells surround the oocyte and mediate follicle growth, estrogen synthesis and ovulatory maturation in response to FSH and LH. In PCOS, granulosa cells often exhibit reduced sensitivity to FSH and dysregulated expression of key factors such as anti-Müllerian hormone (AMH) and its receptor, leading to impaired follicle maturation and ovulatory dysfunction [123,124]. Functional dissection of these pathways using CRISPR-based models has revealed important points of dysregulation in granulosa cell signaling [125].
Recent studies also suggest that A-to-I RNA editing contributes to the pathogenesis of PCOS, particularly through its effects on granulosa cell function [126,127]. In a comprehensive transcriptomic analysis using public datasets from multiple tissues and patient-derived peripheral blood samples, several differentially RNA-edited (DRE) events were consistently identified in PCOS. These events were enriched in genes involved in metabolism, immune regulation, and cell cycle control, including DEGS1, TUBA1B, TRIM56, STK4, ERCC2 and ZNF83 [126,128,129]. While these editing events have not been fully functionally validated in vivo, their recurrence underscores their potential relevance, and species-specific differences in RNA editing must be considered when extrapolating from animal models.
Functional enrichment analysis of these DRE-associated genes revealed significant involvement in immune and apoptotic pathways, including NF-κB signaling and NOD-like receptor signaling, suggesting a potential role for RNA editing in modulating granulosa cell fate and inflammatory responses in PCOS [37,126,130,131]. These findings are consistent with previously reported dysregulation of PI3K/Akt signaling, oxidative stress responses, and miRNA pathways in granulosa cell apoptosis and abnormal follicular development in PCOS [132,133,134].
RNA-binding proteins (RBPs) may also play a role in regulating these RNA editing events. HNRNPDL, in particular, was predicted to bind multiple DRE sites and is known to be involved in transcriptional regulation and alternative splicing [135,136,137]. Its potential involvement in granulosa cell gene regulation in PCOS represents an important avenue for further study. Among RNA editing enzymes, ADAR has been implicated in multiple biological processes and disease states, but its role in PCOS remains poorly defined [138]. One key ADAR target identified in the study was EIF2AK2, which encodes protein kinase R (PKR), a critical component of the innate immune response. EIF2AK2 was consistently differentially edited in both peripheral blood samples and public PCOS datasets [138,139,140]. This gene is involved in the cellular stress response and antiviral defense by regulating mRNA translation and activating downstream inflammatory pathways. In PCOS, EIF2AK2 may act through the MAPK signaling cascade, which has been shown to influence granulosa cell apoptosis and steroid biosynthesis [141,142,143]. The EIF2AK2–MAPK axis could represent a mechanistic link between RNA editing and granulosa cell dysfunction in PCOS.
AMH, secreted by granulosa cells of preantral and small antral follicles, plays a central role in restraining follicular recruitment and modulating FSH sensitivity [144]. Both AMH and its receptor, AMHR2, are elevated in PCOS ovaries and are thought to contribute to follicular arrest [145]. A CRISPR-mediated deletion of the GATA-binding site in the mouse AMH promoter abolished testicular AMH expression without affecting ovarian AMH, demonstrating sexually dimorphic regulatory mechanisms [146]. Although adult female ovaries remained intact, this model confirmed the feasibility of dissecting AMH regulation in vivo [146,147].
A broader approach involving a genome-wide CRISPR knockout screen in sheep granulosa cells cultured with FSH identified over 800 candidate genes implicated in FSH responsiveness [148]. These included multiple epigenetic and transcriptional regulators of FSHR and luteinizing hormone receptor (LHR), expanding the known regulatory landscape beyond canonical FSH signaling and offering a reservoir of candidate genes for future functional studies [148]. In goats (Capra hircus), targeted CRISPR knockout of BMPR1B, a receptor involved in the BMP signaling pathway and mutated in high-fecundity sheep (FecB), disrupted granulosa cell proliferation and steroid hormone production [149]. Loss of BMPR1B enhanced SMAD signaling and upregulated FSHR and LHR expression, indicating altered gonadotropin sensitivity [149]. This model recapitulated aspects of the granulosa cell dysfunction observed in PCOS and further established a role for BMP signaling in granulosa cell competence and ovarian physiology. Together, these studies demonstrate that granulosa cell function in PCOS is shaped by complex interactions between hormone signaling, RNA editing and epigenetic regulation. CRISPR-based functional genomics, combined with emerging insights into RNA-level regulation, offer powerful tools to unravel the molecular basis of granulosa cell dysfunction and identify potential therapeutic targets for PCOS.

3.3. Folliculogenesis and Ovulatory Dysfunction

Folliculogenesis encompasses the sequential maturation of ovarian follicles from primordial through antral stages, culminating in ovulation [150,151]. This process requires coordinated signaling between granulosa and theca cells, estrogen biosynthesis, and extracellular matrix (ECM) remodeling [151,152]. In PCOS, this tightly regulated sequence is disrupted, leading to follicular arrest and anovulation [153].
CRISPR models targeting genes involved in these pathways have demonstrated many of the structural and functional hallmarks of PCOS ovaries [154]. In zebrafish, CRISPR knockout of CYP19A1A abolished estrogen production, resulting in complete ovary-to-testis sex reversal and infertility [155,156]. This extreme phenotype illustrates the essential role of estrogen in establishing and maintaining female reproductive architecture and follicle progression [155,156]. Additionally, CRISPR deletion of ADAMTS9, an ECM protease required for follicle rupture, similarly produced a profound anovulatory phenotype [157]. Female adamts9−/− zebrafish had underdeveloped ovaries composed entirely of early-stage oocytes and failed to ovulate [157,158]. These results confirm ADAMTS9 as a key effector of follicle release and oocyte maturation.
To further probe the balance between inhibitory and stimulatory follicular signals, Wu et al. performed a double knockout of CYP19A1A and AMH [159]. Surprisingly, AMH deletion partially rescued the ovarian phenotype of estrogen-deficient zebrafish, enabling some female differentiation and folliculogenesis [159,160]. This highlights the suppressive effect of AMH on follicle recruitment and its potential role in PCOS pathogenesis when elevated.
Finally, the inducible overexpression of CYP17A1 in theca cells (as described above) provides a mechanistic link between local androgen excess and cyst formation, demonstrating that endocrine cues alone can recapitulate ovulatory dysfunction in the absence of exogenous hormonal induction [115,119,161]. The CRISPR-based gene models described above illustrate the intricate molecular interplay between granulosa and theca cells in ovulatory dysfunction, as summarized in Figure 2.

4. CRISPR in Metabolic Gene Function in PCOS

4.1. Insulin Signaling Pathways

Using pooled and arrayed CRISPR/Cas9 loss-of-function screens, studies in human adipocyte models have shown that knocking out IRS1 causally impairs insulin signaling, evidenced by the reduced insulin-stimulated AKT2 phosphorylation and glucose uptake. In addition, lipid metabolism is modified, thereby establishing a direct role of IRS1 in cellular IR relevant to PCOS metabolic features [162].
CRISPR-based studies that directly target INSR in human adipocytes are limited, and much of our current understanding of INSR and AKT2 in glucose regulation comes from traditional gene-targeted mouse models. For instance, tissue-specific INSR knockouts in mice demonstrate that the insulin receptor has an essential role in maintaining glucose homeostasis [163]. Similarly, mice deficient in AKT2 exhibited hyperglycemia and IR, underscoring AKT2’s crucial role in metabolic control [164]. Likewise, murine embryonic fibroblasts that lack both IRS1 and IRS2 fail to differentiate into adipocytes, showcasing their important role in driving adipogenic transcription factors such as peroxisome proliferator-activated receptor gamma (PPARγ) and C/EBPα [165]. These findings illustrate the mechanistic link between candidate genes and metabolic phenotypes in PCOS, reinforcing their research utility rather than suggesting that gene mutations directly cause the syndrome.

4.2. Adipogenesis and Lipid Regulation

In human preadipocytes, CRISPR/Cas9 knockout of PPARγ prevents differentiation into adipocytes, demonstrating the essential role of PPARγ in regulating adipogenesis [166]. In addition to controlling cell lineage, CRISPR/Cas9 editing of the FTO rs1421085 variant in primary human preadipocytes restored the program for mitochondrial thermogenesis and reversed the obesity-associated shift toward the differentiation of white adipocytes, thereby establishing a causal link between this common variant and adipocyte functional reprogramming [167]. The application of CRISPR has also clarified neuroendocrine drivers of weight that influence the severity of PCOS. In mice, CRISPR knockouts of the LEPR resulted in hyperphagia, obesity, hyperglycemia, and IR [168]. Likewise, a study by Bao et al. generated LEPR-deficient rats [169]. These knockout rats displayed hyperphagia, obesity, dyslipidemia, glucose intolerance, hyperglycemia, and hyperinsulinemia [169]. This reinforces the evolutionary conservation of LEPR function while offering a robust model to explore PCOS-related metabolic disturbances [169]. A study by Hao et al. investigated CRISPR-mediated knockout of the MC4R gene in miniature pigs [170]. Results showed that MC4R-deficient pigs developed marked hyperinsulinemia, IR, hyperorexia, and dysregulated lipid metabolism with substantial hepatic fat accumulation [170]. Moreover, the loss of MC4R resulted in hyperphagia and increased adiposity, culminating in hepatic steatosis even in the absence of an atherogenic diet [170]. This reinforces that obesity-linked metabolic circuits mediated through the melanocortin pathway are evolutionarily conserved across species, hence making them relevant to the metabolic burden often seen in PCOS [170]. These metabolic CRISPR models and their effects are illustrated in Figure 3.

5. Combined Models: Reproductive and Metabolic Phenotypes

Single-gene models have frequently yielded context-dependent or non-translatable phenotypes that complicate inference for human PCOS. For example, FOXL2 loss in adult mice causes granulosa to sertoli transdifferentiation and ovarian sex-reprogramming; an extreme, species-specific outcome that models ovarian failure rather than the chronic endocrine/metabolic dysregulation of PCOS [171]. Androgen receptor (AR) manipulations produce divergent reproductive and metabolic effects depending on whether AR is knocked out or overexpressed and on the developmental window targeted, underscoring sensitivity to timing and modality [172]. Likewise, oocyte-derived factors such as BMP15/GDF9 show marked species differences. Alleles that alter ovulation rate or fertility in sheep or rodents do not consistently reproduce human PCOS features, limiting cross-species extrapolation [173,174]. These discordant outcomes illustrate why single-gene edits can be misleading and strengthen the rationale for dual/multi-gene, temporally controlled, and cross-species CRISPR approaches to model the polygenic, context-dependent nature of human PCOS.

5.1. Dual Gene Models

Dual gene models use CRISPR gene editing to simultaneously target two genes, providing a way to investigate how multiple genetic pathways interact to produce the complex features of a disease [175]. Since PCOS involves both metabolic dysfunction, such as insulin resistance and dyslipidemia, as well as reproductive dysfunction, such as hyperandrogenism and anovulation, dual gene models can offer a more comprehensive insight into the combined effects of these pathways [176]. For instance, CRISPR can be used to edit the reproductive gene DENND1A, which is associated with androgen biosynthesis and elevated testosterone levels, while also targeting a metabolic gene, such as IRS1, which plays a role in insulin signaling and has been found to be impaired in PCOS patients [177,178]. However, despite this theoretical advantage, there is a gap in the literature, where there are no current studies involving dual gene CRISPR models specific to PCOS. This highlights a significant limitation in the field, as understanding how different genes interact may reveal mechanisms that contribute to the complexity of PCOS.
Due to the absence of dual gene models, most studies have focused on a single-gene editing approach. Interestingly, some of these models displayed both reproductive and metabolic phenotypes. For example, a study by Yan et al. (2017) created gonadal soma-derived factor (GSDF) knockout mutations in zebrafish [179]. The mutant females exhibited PCOS features including infertility, oligo-ovulation, hyperandrogenism, IR, and obesity that mimic PCOS, allowing this study to be used as a model to screen for therapies [179]. While not a dual-gene model, this study demonstrates the effect that a single gene knockout had in triggering both reproductive and metabolic dysfunction, highlighting the potential value of future CRISPR-based dual gene models that target ovarian and metabolic genes simultaneously in investigating the gene-gene interactions that contribute to the pathogenesis of PCOS and providing a more accurate representation of the syndrome.

5.2. Integration with Hormonal Induction Models

Traditional animal models of PCOS typically involve induction with letrozole, an aromatase inhibitor, or dehydroepiandrosterone (DHEA) to mimic hyperandrogenism and reproductive dysfunction. Supplementing these protocols with a high-fat diet causes these hormonal models to also develop significant metabolic features, particularly obesity and IR. For instance, a rat model treated with both letrozole and a high-fat diet displayed anovulatory cycles, polycystic ovaries, elevated testosterone, glucose intolerance and dyslipidemia, along with impaired insulin signaling, as evidenced by reduced phosphorylation of INSR, IRS, PI3K and AKT in classic insulin-target tissues [180]. Similarly, combining a high-fat diet with DHEA in mice resulted in changes that mimicked those of clinical PCOS. It showed dyslipidemia, elevated LH and serum testosterone, development of multiple ovarian cysts and poor ovarian microenvironment, features that were less pronounced with letrozole or DHEA alone [181].
Although both CRISPR-mediated knockouts of reproductive or metabolic genes and hormone-induced PCOS models (such as letrozole or DHEA induction) are commonly used, a combined approach integrating gene editing with hormonal induction models remains unexplored in the literature. Such combined designs, where CRISPR-modified rodents are further treated with letrozole or DHEA, could provide powerful insight into gene-environment interactions that better replicate the dual metabolic and reproductive phenotypes seen in women with PCOS. Integrating CRISPR gene edits with hormonal induction allows mechanistic exploration of gene-environment interactions, providing insight into human disease pathways without implying immediate clinical application. These models can also further offer a more robust platform for testing therapeutic interventions, as drug responses can be assessed in the presence of both genetic susceptibility and hormonal imbalance, thereby reflecting human disease conditions more accurately. However, to date, no studies have implemented or reported such integrated models.

6. Insights from CRISPR Models

6.1. Validation of Candidate Gene Roles

While GWAS has successfully identified numerous loci associated with PCOS, these associations alone do not prove that these genes are truly involved in the pathogenesis of the disease [182]. To bridge this gap, CRISPR-based gene editing can directly target the genes, allowing researchers to assess their role in causing the PCOS-relevant phenotypes. For example, CRISPR/Cas9 was used to generate LNK (SH2B3) knockout mice, who were fed with a high-fat diet and injected with DHEA to induce PCOS features [183]. Since LNK is a crucial regulator of the insulin signaling pathway, the study aimed to investigate its effect on the pathogenesis of PCOS [183]. Compared to wild-type PCOS mice, the LNK knockout group exhibited a partially restored estrous cycle and an improved glucose metabolism, indicating that LNK may be a target for PCOS clinical treatment [183]. Therefore, the progression from GWAS-only associations to CRISPR-based functional confirmation highlights its potential in identifying genes that play a direct role in PCOS development, helping researchers move from correlation to causation.

6.2. Novel Discoveries from Loss-of-Function or Gain-of-Function Studies

Beyond confirming candidate gene roles, CRISPR has also emerged as a discovery tool, uncovering unexpected ways that certain genes behave in PCOS. By enabling targeted loss- or gain-of-function, CRISPR can challenge previous assumptions about a gene’s involvement in PCOS pathophysiology. For example, showing that a gene strongly associated with PCOS in GWAS may have no significant effect in knockout models, while others may unexpectedly drive disease features when overexpressed. Such findings are especially important in a complex condition like PCOS, where the overlapping of reproductive, metabolic, and other pathways provides outcomes that are not always predictable.
For instance, the THADA gene encodes thyroid adenoma-associated protein, which is expressed in various tissues, including the ovaries [184]. Studies have revealed its association with disruptions in energy metabolism, increasing susceptibility to obesity and the likelihood of developing PCOS [185]. Additionally, GWASs have identified THADA as a candidate gene for PCOS [184]. Therefore, an animal study aimed to prove that THADA had an effect in vivo on the female reproductive system, using CRISPR/Cas9 technology to create THADA knockout mice [184]. Surprisingly, this study showed that THADA ablation did not result in the expected abnormalities, given that the knockout mice had normal fertility, no significant differences in body weight compared to the wild-type mice and no ovarian dysfunction [184]. The study further tested the influence of environmental factors, specifically dietary factors, on PCOS development, and despite stressing the mice with a high-fat-high-sugar (HFHS) diet, they still exhibited a relatively stable ovarian function [184]. This ultimately demonstrates CRISPR’s role in reshaping our understanding of PCOS and shifts our focus to other tissues or pathways.
Recently, a study utilized CRISPR/Cas9 technology to test the involvement of IGFBP7, a gene that produces the glycoprotein Insulin-like Growth Factor Binding Protein 7. It is known for regulating vascular remodeling and modulating the pro-angiogenic properties of signaling factors like VEGF-A and IGF [186]. Knowing that IGFBP7 plays a role in regulating immune dysfunction, its expression levels were tested in PCOS patients and found to be significantly upregulated [187]. Therefore, an animal-based model was created to explore its involvement in immune modulation within the context of PCOS [187]. Briefly, CRISPR was used to create IGFBP7 knockout mice, and DHEA was administered daily for 21 days to both the knockout and wild-type groups to induce PCOS-like symptoms [187]. As a result, the wild-type mice exhibited features of PCOS, including disruption of their estrous cycle, while the knockout group did not display such features, indicating that turning off the IGFBP7 gene reversed the disruption caused by the administered DHEA [187]. Furthermore, the ovaries of the IGFBP7+/+NC group exhibited cystic follicular enlargement, reduced granulosa cell layers, a great number of immature and atretic follicles and the disappearance of oocytes [187]. Despite being under-researched in the context of PCOS, this study reveals that IGFBP7 plays a crucial role in the development of PCOS and is worthy of further exploration. Key CRISPR-based loss- and gain-of-function studies investigating PCOS-associated genes are summarized in Table 2.

7. Future Directions and Clinical Translation

Although GWASs have uncovered numerous genetic loci associated with PCOS, functional validation of these candidates remains incomplete [188]. CRISPR-based technologies have emerged as powerful tools to interrogate gene function with unprecedented precision, enabling targeted manipulation of PCOS-associated genes across ovarian and metabolic tissues [189]. However, translating these advances into clinically meaningful insights requires overcoming key limitations in current models, namely, species-specific differences in ovarian physiology, limited phenotypic fidelity and insufficient temporal control [190]. To shift from gene identification toward therapeutic translation, future research must prioritize two key strategies: (1) integration of CRISPR with multi-omic platforms and (2) precision gene editing tailored to molecularly defined PCOS subtypes.

7.1. Integrating CRISPR with Multi-Omic Frameworks

PCOS is a multifactorial disorder involving gene regulation across ovarian, hypothalamic, and metabolic tissues [45]. Integrating CRISPR perturbations with multi-omics, such as bulk and single-cell RNA-seq, ATAC-seq, ChIP-seq, and proteomics, can capture the multilayered consequences of gene editing in a tissue-specific manner. For example, CRISPR-mediated knockout of DENND1A.V2 in primary theca cells or steroidogenic cell lines [118] could be coupled with single-cell transcriptomics to identify downstream effectors of androgen synthesis [118,191], while concurrent ATAC-seq could reveal changes in chromatin accessibility at promoters and enhancers governing steroidogenesis [192]. Similarly, base editing of regulatory SNPs within PCOS-associated enhancers (e.g., near FSHR, GATA4 or LHCGR) in granulosa-like induced pluripotent stem cell (iPSC)-derived cells can provide direct evidence of variant-driven transcriptional modulation [193,194,195]. In vivo, conditional CRISPR models integrated with spatial transcriptomics or single-cell multi-omic profiling in mouse ovaries could clarify how gene perturbations reshape cellular niches and intercellular signaling during folliculogenesis. These strategies will allow researchers to map the effect of a given gene, as well as its dynamic regulatory network across cell types and time points.

7.2. Precision Editing in PCOS

A major barrier to clinical translation is the heterogeneity of PCOS phenotypes [196]. Subgroup-specific genetic dependencies, such as elevated androgen biosynthesis in “hyperandrogenic PCOS” or insulin resistance in “metabolic PCOS,” suggest that therapeutic CRISPR strategies must be personalized [197]. In the near-term, CRISPR-derived tools such as dCas9-fused repressors (CRISPRi) [198,199] or epigenetic silencers (e.g., KRAB, HDACs) can be adapted to transiently modulate genes like CYP17A1 or AMHR2 in ovarian cells, without inducing double-strand breaks. This may reduce off-target risks while permitting reversibility, which is ideal for cyclic or temporally restricted pathologies such as anovulation.
Emerging delivery systems such as lipid nanoparticles, viral vectors with tissue-specific promoters, and engineered extracellular vesicles offer potential routes for delivering CRISPR-based therapies to ovaries, liver or adipose tissue [200,201]. For instance, targeted downregulation of INSR inhibitors or reactivation of dormant FSHR pathways via CRISPRa in granulosa cells could normalize ovulatory cycles in selected patient subtypes [202].
In the long term, integrating patient-specific genomic data with gene editing holds promise for personalized medicine. PCOS organoids and iPSC-derived follicular structures generated from patient cells can serve as preclinical platforms for screening CRISPR-based interventions, toxicity, and efficacy before translation to clinical trials [203,204]. Germline editing remains inappropriate for PCOS applications, but somatic and ex vivo approaches, particularly in reproductive tissues, may enter therapeutic development once safety, efficiency, and reversibility are established [205,206].

7.3. Safety, Delivery and Ethical Considerations

Clinical translation of CRISPR for PCOS requires an integrated approach addressing safety, off-target effects, targeted delivery, temporal control, and ethical oversight. Off-target cleavage, large deletions, chromosomal rearrangements, p53-mediated responses, and immune activation are key safety concerns [207,208]. High-fidelity Cas variants (eSpCas9, HiFi Cas9, HypaCas9), base editors, and prime editors significantly reduce these risks by enhancing target specificity and reducing unintended genome modifications [209]. Genome-wide off-target detection methods such as GUIDE-seq, CIRCLE-seq, and Digenome-seq remain essential for preclinical validation [210,211]. Efficient delivery remains critical, with viral vectors (AAV, lentivirus) and non-viral platforms (lipid nanoparticles, polymeric carriers, engineered extracellular vesicles) enabling transient, tissue-specific targeting of ovaries, liver, or adipose tissue while minimizing immunogenicity and genotoxicity [212]. Preclinical evaluation of biodistribution, pharmacokinetics, and large-animal ovarian models is recommended to ensure translational safety [213,214]. Temporal control is particularly important in cyclic ovarian physiology; inducible CRISPR systems, RNP/mRNA delivery, CRISPRi/CRISPRa, and anti-CRISPR proteins allow reversible modulation to prevent long-term adverse effects [215,216]. Ethical and regulatory considerations require balancing permissible somatic applications with prohibitions on germline editing [217,218], while addressing informed consent, intergenerational impact, equitable access, and compliance with national and international guidelines. Long-term monitoring for safety, efficacy, and societal impact is critical, and integrating these strategies with multi-omic and precision editing platforms provides a comprehensive roadmap for responsible CRISPR translation in PCOS research.

8. Conclusions

CRISPR technology has provided a turning point in PCOS research, enabling functional validation of candidate genes and clarifying the mechanisms that connect reproductive and metabolic dysfunction. Although most studies still rely on single-gene models, the integration of dual-gene and hormone induction models, patient-derived organoids and multi-omic profiling will likely be necessary to reflect the full complexity of the syndrome. CRISPR not only enhances our understanding of the mechanisms of PCOS, but also sets the foundation for developing precise, gene-targeted therapies.

Author Contributions

Conceptualization, S.B. (Shahd Bucheeri) and Y.A.; methodology, S.B. (Shahd Bucheeri) and Y.A.; investigation, S.B. (Shahd Bucheeri), Y.A., Y.B., S.B. (Sarah Bucheeri) and A.A.; writing—original draft preparation, S.B. (Shahd Bucheeri), Y.A., Y.B., S.B. (Sarah Bucheeri). and A.A.; writing—review and editing, S.B. (Shahd Bucheeri), Y.A., Y.B., S.B. (Sarah Bucheeri), A.A. and A.E.B.; visualization, S.B. (Shahd Bucheeri), Y.A., Y.B. and S.B. (Sarah Bucheeri); A.E.B. is the guarantor of this work. All authors have read and agreed to the published version of the manuscript.

Funding

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

No authors have any competing interests to declare, financial or otherwise.

References

  1. Siddiqui, S.; Mateen, S.; Ahmad, R.; Moin, S. A brief insight into the etiology, genetics, and immunology of polycystic ovarian syndrome (PCOS). J. Assist. Reprod. Genet. 2022, 39, 2439–2473. [Google Scholar] [CrossRef]
  2. Sidra, S.; Tariq, M.H.; Farrukh, M.J.; Mohsin, M. Evaluation of clinical manifestations, health risks, and quality of life among women with polycystic ovary syndrome. PLoS ONE 2019, 14, e0223329. [Google Scholar] [CrossRef]
  3. Chen, Y.; Ma, L.; Ge, Z.; Pan, Y.; Xie, L. Key Genes Associated With Non-Alcoholic Fatty Liver Disease and Polycystic Ovary Syndrome. Front. Mol. Biosci. 2022, 9, 888194. [Google Scholar] [CrossRef]
  4. Hoeger, K.M.; Dokras, A.; Piltonen, T. Update on PCOS: Consequences, Challenges, and Guiding Treatment. J. Clin. Endocrinol. Metab. 2020, 106, e1071–e1083. [Google Scholar] [CrossRef]
  5. Joshi, A. PCOS stratification for precision diagnostics and treatment. Front. Cell Dev. Biol. 2024, 12, 1358755. [Google Scholar] [CrossRef] [PubMed]
  6. Mikhael, S.; Punjala-Patel, A.; Gavrilova-Jordan, L. Hypothalamic-Pituitary-Ovarian Axis Disorders Impacting Female Fertility. Biomedicines 2019, 7, 5. [Google Scholar] [CrossRef] [PubMed]
  7. Christian, C.A.; Moenter, S.M. The Neurobiology of Preovulatory and Estradiol-Induced Gonadotropin-Releasing Hormone Surges. Endocr. Rev. 2010, 31, 544–577. [Google Scholar] [CrossRef] [PubMed]
  8. Taylor, A.E.; McCourt, B.; Martin, K.A.; Anderson, E.J.; Adams, J.M.; Schoenfeld, D.; Hall, J.E. Determinants of Abnormal Gonadotropin Secretion in Clinically Defined Women with Polycystic Ovary Syndrome1. J. Clin. Endocrinol. Metab. 1997, 82, 2248–2256. [Google Scholar] [CrossRef]
  9. Zhong, Y.; Ding, J.; Xia, F. DNA methylation and its impact on ovarian steroidogenesis in women with polycystic ovary syndrome: Insights from human and animal models. Arch. Gynecol. Obstet. 2025, 312, 363–374. [Google Scholar] [CrossRef]
  10. Zheng, M.; Andersen, C.Y.; Rasmussen, F.R.; Cadenas, J.; Christensen, S.T.; Mamsen, L.S. Expression of genes and enzymes involved in ovarian steroidogenesis in relation to human follicular development. Front. Endocrinol. 2023, 14, 1268248. [Google Scholar] [CrossRef]
  11. Rosenfield, R.L.; Ehrmann, D.A. The Pathogenesis of Polycystic Ovary Syndrome (PCOS): The Hypothesis of PCOS as Functional Ovarian Hyperandrogenism Revisited. Endocr. Rev. 2016, 37, 467–520. [Google Scholar] [CrossRef]
  12. Panghiyangani, R.; Soeharso, P.; Andrijono; Suryandari, D.A.; Wiweko, B.; Kurniati, M.; Pujianto, D.A. CYP19A1 gene expression in patients with polycystic ovarian syndrome. J. Hum. Reprod. Sci. 2020, 13, 100–103. [Google Scholar] [CrossRef]
  13. Azhary, J.M.K.; Harada, M.; Kunitomi, C.; Kusamoto, A.; Takahashi, N.; Nose, E.; Oi, N.; Wada-Hiraike, O.; Urata, Y.; Hirata, T.; et al. Androgens Increase Accumulation of Advanced Glycation End Products in Granulosa Cells by Activating ER Stress in PCOS. Endocrinology 2020, 161, bqaa015. [Google Scholar] [CrossRef]
  14. Liao, B.; Qi, X.; Yun, C.; Qiao, J.; Pang, Y. Effects of Androgen Excess-Related Metabolic Disturbances on Granulosa Cell Function and Follicular Development. Front. Endocrinol. 2022, 13, 815968. [Google Scholar] [CrossRef] [PubMed]
  15. Wu, Y.; Li, Y.; Liao, X.; Wang, Z.; Li, R.; Zou, S.; Jiang, T.; Zheng, B.; Duan, P.; Xiao, J. Diabetes Induces Abnormal Ovarian Function via Triggering Apoptosis of Granulosa Cells and Suppressing Ovarian Angiogenesis. Int. J. Biol. Sci. 2017, 13, 1297–1308. [Google Scholar] [CrossRef] [PubMed]
  16. Harada, M.; Nose, E.; Takahashi, N.; Hirota, Y.; Hirata, T.; Yoshino, O.; Koga, K.; Fujii, T.; Osuga, Y. Evidence of the activation of unfolded protein response in granulosa and cumulus cells during follicular growth and maturation. Gynecol. Endocrinol. 2015, 31, 783–787. [Google Scholar] [CrossRef]
  17. Park, H.; Park, J.; Kim, J.; Yang, S.; Jung, J.; Kim, M.; Kang, M.; Cho, Y.H.; Wee, G.; Yang, H.; et al. Melatonin improves the meiotic maturation of porcine oocytes by reducing endoplasmic reticulum stress during in vitro maturation. J. Pineal Res. 2017, 64, e12458. [Google Scholar] [CrossRef]
  18. Sørensen, A.E.; Udesen, P.B.; Wissing, M.L.; Englund, A.L.M.; Dalgaard, L.T. MicroRNAs related to androgen metabolism and polycystic ovary syndrome. Chem. Interact. 2016, 259, 8–16. [Google Scholar] [CrossRef]
  19. Kahn, S.E.; Hull, R.L.; Utzschneider, K.M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 2006, 444, 840–846. [Google Scholar] [CrossRef]
  20. Cara, J.F.; Rosenfield, R.L. Insulin-Like Growth Factor I and Insulin Potentiate Luteinizing Hormone-Induced Androgen Synthesis by Rat Ovarian Thecal-Interstitial Cells. Endocrinology 1988, 123, 733–739. [Google Scholar] [CrossRef]
  21. Zhang, G.; Garmey, J.C.; Veldhuis, J.D. Interactive Stimulation by Luteinizing Hormone and Insulin of the Steroidogenic Acute Regulatory (StAR) Protein and 17α-Hydroxylase/17, 20-Lyase (CYP17) Genes in Porcine Theca Cells. Endocrinology 2000, 141, 2735–2742. [Google Scholar] [CrossRef]
  22. Toprak, S.; Yönem, A.; Çakır, B.; Güler, S.; Azal, Ö.; Özata, M.; Çorakçı, A. Insulin Resistance in Nonobese Patients with Polycystic Ovary Syndrome. Horm. Res. Paediatr. 2001, 55, 65–70. [Google Scholar] [CrossRef]
  23. Bulsara, J.; Patel, P.; Soni, A.; Acharya, S. A review: Brief insight into Polycystic Ovarian syndrome. Endocr. Metab. Sci. 2021, 3, 100085. [Google Scholar] [CrossRef]
  24. Goodarzi, M.O.; Dumesic, D.A.; Chazenbalk, G.; Azziz, R. Polycystic ovary syndrome: Etiology, pathogenesis and diagnosis. Nat. Rev. Endocrinol. 2011, 7, 219–231. [Google Scholar] [CrossRef]
  25. Rojas, J.; Chávez, M.; Olivar, L.; Rojas, M.; Morillo, J.; Mejías, J.; Calvo, M.; Bermúdez, V. Polycystic Ovary Syndrome, Insulin Resistance, and Obesity: Navigating the Pathophysiologic Labyrinth. Int. J. Reprod. Med. 2014, 2014, 719050. [Google Scholar] [CrossRef] [PubMed]
  26. Dunaif, A. Insulin Resistance and the Polycystic Ovary Syndrome: Mechanism and Implications for Pathogenesis. Endocr. Rev. 1997, 18, 774–800. [Google Scholar] [CrossRef]
  27. Teede, H.J.; Tay, C.T.; Laven, J.; Dokras, A.; Moran, L.J.; Piltonen, T.T.; Costello, M.F.; Boivin, J.; Redman, L.M.; Boyle, J.A.; et al. Recommendations from the 2023 International Evidence-based Guideline for the Assessment and Management of Polycystic Ovary Syndrome. Fertil. Steril. 2023, 120, 767–793. [Google Scholar] [CrossRef]
  28. Ding, H.; Zhang, J.; Zhang, F.; Zhang, S.; Chen, X.; Liang, W.; Xie, Q. Resistance to the Insulin and Elevated Level of Androgen: A Major Cause of Polycystic Ovary Syndrome. Front. Endocrinol. 2021, 12, 741764. [Google Scholar] [CrossRef]
  29. Marshall, J.C.; Dunaif, A. Should all women with PCOS be treated for insulin resistance? Fertil. Steril. 2012, 97, 18–22. [Google Scholar] [CrossRef]
  30. De Leo, V.; la Marca, A.; Petraglia, F. Insulin-Lowering Agents in the Management of Polycystic Ovary Syndrome. Endocr. Rev. 2003, 24, 633–667. [Google Scholar] [CrossRef]
  31. Kelly, C.C.J.; Lyall, H.; Petrie, J.R.; Gould, G.W.; Connell, J.M.C.; Sattar, N. Low Grade Chronic Inflammation in Women with Polycystic Ovarian Syndrome. J. Clin. Endocrinol. Metab. 2001, 86, 2453–2455. [Google Scholar] [CrossRef]
  32. Orio, F.; Palomba, S.; Cascella, T.; Di Biase, S.; Manguso, F.; Tauchmanovà, L.; Nardo, L.G.; Labella, D.; Savastano, S.; Russo, T.; et al. The Increase of Leukocytes as a New Putative Marker of Low-Grade Chronic Inflammation and Early Cardiovascular Risk in Polycystic Ovary Syndrome. J. Clin. Endocrinol. Metab. 2005, 90, 2–5. [Google Scholar] [CrossRef] [PubMed]
  33. Phelan, N.; O’COnnor, A.; Tun, T.K.; Correia, N.; Boran, G.; Roche, H.M.; Gibney, J. Leucocytosis in women with polycystic ovary syndrome (PCOS) is incompletely explained by obesity and insulin resistance. Clin. Endocrinol. 2012, 78, 107–113. [Google Scholar] [CrossRef] [PubMed]
  34. Escobar-Morreale, H.F.; Botella-Carretero, J.I.; Villuendas, G.; Sancho, J.; Millán, J.L.S. Serum Interleukin-18 Concentrations Are Increased in the Polycystic Ovary Syndrome: Relationship to Insulin Resistance and to Obesity. J. Clin. Endocrinol. Metab. 2004, 89, 806–811. [Google Scholar] [CrossRef]
  35. Thathapudi, S.; Kodati, V.; Erukkambattu, J.; Katragadda, A.; Addepally, U.; Hasan, Q. Tumor Necrosis Factor-Alpha and Polycystic Ovarian Syndrome: A Clinical, Biochemical, and Molecular Genetic Study. Genet. Test. Mol. Biomark. 2014, 18, 605–609. [Google Scholar] [CrossRef] [PubMed]
  36. Sadeghi, A.; Fadaei, R.; Moradi, N.; Fouani, F.Z.; Roozbehkia, M.; Zandieh, Z.; Ansaripour, S.; Vatannejad, A.; Doustimotlagh, A.H. Circulating levels of C1q/TNF-α-related protein 6 (CTRP6) in polycystic ovary syndrome. IUBMB Life 2020, 72, 1449–1459. [Google Scholar] [CrossRef]
  37. Yan, S.; Ding, J.; Zhang, Y.; Wang, J.; Zhang, S.; Yin, T.; Yang, J. C1QTNF6 participates in the pathogenesis of PCOS by affecting the inflammatory response of granulosa cells. Biol. Reprod. 2021, 105, 427–438. [Google Scholar] [CrossRef]
  38. Gonzalez, F.; Thusu, K.; Abdel-Rahman, E.; Prabhala, A.; Tomani, M.; Dandona, P. Elevated serum levels of tumor necrosis factor alpha in normal-weight women with polycystic ovary syndrome. Metabolism 1999, 48, 437–441. [Google Scholar] [CrossRef]
  39. Sayin, N.C.; Gücer, F.; Balkanli-Kaplan, P.; Yüce, M.A.; Ciftci, S.; Kücük, M.; Yardim, T. Elevated serum TNF-alpha levels in normal-weight women with polycystic ovaries or the polycystic ovary syndrome. J. Reprod. Med. 2003, 48, 165–170. [Google Scholar]
  40. Amato, G.; Conte, M.; Mazziotti, G.; Lalli, E.; Vitolo, G.; Tucker, A.T.; Bellastella, A.; Carella, C.; Izzo, A. Serum and follicular fluid cytokines in polycystic ovary syndrome during stimulated cycles. Obstet. Gynecol. 2003, 101, 1177–1182. [Google Scholar] [CrossRef]
  41. Knebel, B.; Janssen, O.; Hahn, S.; Jacob, S.; Gleich, J.; Kotzka, J.; Muller-Wieland, D. Increased Low Grade Inflammatory Serum Markers in Patients with Polycystic Ovary Syndrome (PCOS) and their Relationship to PPARγ Gene Variants. Exp. Clin. Endocrinol. Diabetes 2008, 116, 481–486. [Google Scholar] [CrossRef]
  42. Zhang, Y.; Proenca, R.; Maffei, M.; Barone, M.; Leopold, L.; Friedman, J.M. Positional cloning of the mouse obese gene and its human homologue. Nature 1994, 372, 425–432. [Google Scholar] [CrossRef]
  43. Dewailly, D.; Lujan, M.E.; Carmina, E.; Cedars, M.I.; Laven, J.; Norman, R.J.; Escobar-Morreale, H.F. Definition and significance of polycystic ovarian morphology: A task force report from the Androgen Excess and Polycystic Ovary Syndrome Society. Hum. Reprod. Updat. 2013, 20, 334–352. [Google Scholar] [CrossRef]
  44. Diamanti-Kandarakis, E.; Dunaif, A. Insulin Resistance and the Polycystic Ovary Syndrome Revisited: An Update on Mechanisms and Implications. Endocr. Rev. 2012, 33, 981–1030. [Google Scholar] [CrossRef]
  45. Su, P.; Chen, C.; Sun, Y. Physiopathology of polycystic ovary syndrome in endocrinology, metabolism and inflammation. J. Ovarian Res. 2025, 18, 1–10. [Google Scholar] [CrossRef]
  46. Azhary, J.M.K.; Harada, M.; Takahashi, N.; Nose, E.; Kunitomi, C.; Koike, H.; Hirata, T.; Hirota, Y.; Koga, K.; Wada-Hiraike, O.; et al. Endoplasmic Reticulum Stress Activated by Androgen Enhances Apoptosis of Granulosa Cells via Induction of Death Receptor 5 in PCOS. Endocrinology 2018, 160, 119–132. [Google Scholar] [CrossRef] [PubMed]
  47. Phillip, J.; Cartwright, S.; Scott, A. The size and morphology of T.G.E. and vomiting and wasting disease viruses of pigs. Vet. Rec. 1971, 88, 311–312. [Google Scholar] [CrossRef] [PubMed]
  48. Ghafari, A.; Maftoohi, M.; Samarin, M.E.; Barani, S.; Banimohammad, M.; Samie, R. The last update on polycystic ovary syndrome(PCOS), diagnosis criteria, and novel treatment. Endocr. Metab. Sci. 2025, 17, 100228. [Google Scholar] [CrossRef]
  49. Mahoney, A.; D’angelo, A. Treatment Options for Managing Anovulation in Women with PCOS: An Extensive Literature Review of Evidence-Based Recommendations for Future Directions. Life 2025, 15, 863. [Google Scholar] [CrossRef]
  50. Szczesnowicz, A.; Szeliga, A.; Niwczyk, O.; Bala, G.; Meczekalski, B. Do GLP-1 Analogs Have a Place in the Treatment of PCOS? New Insights and Promising Therapies. J. Clin. Med. 2023, 12, 5915. [Google Scholar] [CrossRef]
  51. Fitz, V.; Graca, S.; Mahalingaiah, S.; Liu, J.; Lai, L.; Butt, A.; Armour, M.; Rao, V.; Naidoo, D.; Maunder, A.; et al. Inositol for Polycystic Ovary Syndrome: A Systematic Review and Meta-analysis to Inform the 2023 Update of the International Evidence-based PCOS Guidelines. J. Clin. Endocrinol. Metab. 2024, 109, 1630–1655. [Google Scholar] [CrossRef]
  52. Ma, Y.; Zhang, L.; Huang, X. Genome modification by CRISPR/Cas9. FEBS J. 2014, 281, 5186–5193. [Google Scholar] [CrossRef]
  53. Chen, Y.; Wang, G.; Chen, J.; Wang, C.; Dong, X.; Chang, H.-M.; Yuan, S.; Zhao, Y.; Mu, L. Genetic and Epigenetic Landscape for Drug Development in Polycystic Ovary Syndrome. Endocr. Rev. 2024, 45, 437–459. [Google Scholar] [CrossRef] [PubMed]
  54. Carmina, E. The Role of Gene Alterations in the Pathogenesis of Polycystic Ovary Syndrome. J. Clin. Med. 2025, 14, 3347. [Google Scholar] [CrossRef] [PubMed]
  55. Hayes, M.G.; Urbanek, M.; Ehrmann, D.A.; Armstrong, L.L.; Lee, J.Y.; Sisk, R.; Karaderi, T.; Barber, T.M.; McCarthy, M.I.; Franks, S.; et al. Genome-wide association of polycystic ovary syndrome implicates alterations in gonadotropin secretion in European ancestry populations. Nat. Commun. 2015, 6, 7502. [Google Scholar] [CrossRef] [PubMed]
  56. Dapas, M.; Lin, F.T.J.; Nadkarni, G.N.; Sisk, R.; Legro, R.S.; Urbanek, M.; Hayes, M.G.; Dunaif, A. Distinct subtypes of polycystic ovary syndrome with novel genetic associations: An unsupervised, phenotypic clustering analysis. PLoS Med. 2020, 17, e1003132. [Google Scholar] [CrossRef]
  57. McAllister, J.M.; Modi, B.; Miller, B.A.; Biegler, J.; Bruggeman, R.; Legro, R.S.; Strauss, J.F. Overexpression of a DENND1A isoform produces a polycystic ovary syndrome theca phenotype. Proc. Natl. Acad. Sci. USA 2014, 111, E1519–E1527. [Google Scholar] [CrossRef]
  58. Zeggini, E.; Scott, L.J.; Saxena, R.; Voight, B.F.; Marchini, J.L.; Hu, T.; de Bakker, P.I.; Abecasis, G.R.; Almgren, P.; Andersen, G.; et al. Meta-analysis of genome-wide association data and large-scale replication identifies additional susceptibility loci for type 2 diabetes. Nat. Genet. 2008, 40, 638–645. [Google Scholar] [CrossRef] [PubMed]
  59. Moraru, A.; Cakan-Akdogan, G.; Strassburger, K.; Males, M.; Mueller, S.; Jabs, M.; Muelleder, M.; Frejno, M.; Braeckman, B.P.; Ralser, M.; et al. THADA Regulates the Organismal Balance between Energy Storage and Heat Production. Dev. Cell 2017, 41, 72–81.e6. [Google Scholar] [CrossRef]
  60. Rasool, S.U.A.; Ashraf, S.; Nabi, M.; Masoodi, S.R.; Fazili, K.M.; Amin, S. Clinical Manifestations of Hyperandrogenism and Ovulatory Dysfunction Are Not Associated with His1058 C/T SNP (rs1799817) Polymorphism of Insulin Receptor Gene Tyrosine Kinase Domain in Kashmiri Women with PCOS. Int. J. Endocrinol. 2021, 2021, 1–10. [Google Scholar] [CrossRef]
  61. Singh, R.; Kaur, S.; Yadav, S.; Bhatia, S. Gonadotropins as pharmacological agents in assisted reproductive technology and polycystic ovary syndrome. Trends Endocrinol. Metab. 2023, 34, 194–215. [Google Scholar] [CrossRef] [PubMed]
  62. Valkenburg, O.; Uitterlinden, A.; Piersma, D.; Hofman, A.; Themmen, A.; de Jong, F.; Fauser, B.; Laven, J. Genetic polymorphisms of GnRH and gonadotrophic hormone receptors affect the phenotype of polycystic ovary syndrome. Hum. Reprod. 2009, 24, 2014–2022. [Google Scholar] [CrossRef]
  63. Laven, J.S.; Mulders, A.G.; A Suryandari, D.; Gromoll, J.; Nieschlag, E.; Fauser, B.C.; Simoni, M. Follicle-stimulating hormone receptor polymorphisms in women with normogonadotropic anovulatory infertility. Fertil. Steril. 2003, 80, 986–992. [Google Scholar] [CrossRef]
  64. Farsimadan, M.; Ghosi, F.M.; Takamoli, S.; Vaziri, H. Association analysis of KISS1 polymorphisms and haplotypes with polycystic ovary syndrome. Br. J. Biomed. Sci. 2021, 78, 201–205. [Google Scholar] [CrossRef]
  65. Albalawi, F.S.; Daghestani, M.H.; Daghestani, M.H.; Eldali, A.; Warsy, A.S. rs4889 polymorphism in KISS1 gene, its effect on polycystic ovary syndrome development and anthropometric and hormonal parameters in Saudi women. J. Biomed. Sci. 2018, 25, 50. [Google Scholar] [CrossRef]
  66. Porter, D.T.; Moore, A.M.; A Cobern, J.; Padmanabhan, V.; Goodman, R.L.; Coolen, L.M.; Lehman, M.N. Prenatal Testosterone Exposure Alters GABAergic Synaptic Inputs to GnRH and KNDy Neurons in a Sheep Model of Polycystic Ovarian Syndrome. Endocrinology 2019, 160, 2529–2542. [Google Scholar] [CrossRef]
  67. Abbara, A.; Eng, P.C.; Phylactou, M.; Clarke, S.A.; Richardson, R.; Sykes, C.M.; Phumsatitpong, C.; Mills, E.; Modi, M.; Izzi-Engbeaya, C.; et al. Kisspeptin receptor agonist has therapeutic potential for female reproductive disorders. J. Clin. Investig. 2020, 130, 6739–6753. [Google Scholar] [CrossRef]
  68. Abbara, A.; Ufer, M.; Voors-Pette, C.; Berman, L.; Ezzati, M.; Wu, R.; Lee, T.-Y.; Ferreira, J.C.A.; Migoya, E.; Dhillo, W.S. Endocrine profile of the kisspeptin receptor agonist MVT-602 in healthy premenopausal women with and without ovarian stimulation: Results from 2 randomized, placebo-controlled clinical tricals. Fertil. Steril. 2023, 121, 95–106. [Google Scholar] [CrossRef]
  69. Dallel, M.; Douma, Z.; Finan, R.R.; Hachani, F.; Letaifa, D.B.; Mahjoub, T.; Almawi, W.Y. Contrasting association of Leptin receptor polymorphisms and haplotypes with polycystic ovary syndrome in Bahraini and Tunisian women: A case–control study. Biosci. Rep. 2021, 41, BSR20202726. [Google Scholar] [CrossRef] [PubMed]
  70. Pereira, S.; Cline, D.L.; Glavas, M.M.; Covey, S.D.; Kieffer, T.J. Tissue-Specific Effects of Leptin on Glucose and Lipid Metabolism. Endocr. Rev. 2021, 42, 1–28. [Google Scholar] [CrossRef] [PubMed]
  71. Liang, J.; Lan, J.; Li, M.; Wang, F. Associations of Leptin Receptor and Peroxisome Proliferator-Activated Receptor Gamma Polymorphisms with Polycystic Ovary Syndrome: A Meta-Analysis. Ann. Nutr. Metab. 2019, 75, 1–8. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Q.; Liu, H.; Bai, H.; Huang, W.; Zhang, R.; Tan, J.; Guan, L.; Fan, P. Association of SOD2 A16V and PON2 S311C polymorphisms with polycystic ovary syndrome in Chinese women. J. Endocrinol. Investig. 2019, 42, 909–921. [Google Scholar] [CrossRef]
  73. Bresciani, G.; Cruz, I.B.M.; de Paz, J.A.; Cuevas, M.J.; González-Gallego, J. The MnSOD Ala16Val SNP: Relevance to human diseases and interaction with environmental factors. Free Radic. Res. 2013, 47, 781–792. [Google Scholar] [CrossRef] [PubMed]
  74. Jamnongjit, M.; Gill, A.; Hammes, S.R. Epidermal growth factor receptor signaling is required for normal ovarian steroidogenesis and oocyte maturation. Proc. Natl. Acad. Sci. USA 2005, 102, 16257–16262. [Google Scholar] [CrossRef]
  75. Peng, Y.; Zhang, W.; Yang, P.; Tian, Y.; Su, S.; Zhang, C.; Chen, Z.-J.; Zhao, H. ERBB4 Confers Risk for Polycystic Ovary Syndrome in Han Chinese. Sci. Rep. 2017, 7, srep42000. [Google Scholar] [CrossRef]
  76. Day, F.; Karaderi, T.; Jones, M.R.; Meun, C.; He, C.; Drong, A.; Kraft, P.; Lin, N.; Huang, H.; Broer, L.; et al. Large-scale genome-wide meta-analysis of polycystic ovary syndrome suggests shared genetic architecture for different diagnosis criteria. PLoS Genet. 2018, 14, e1007813. [Google Scholar] [CrossRef] [PubMed]
  77. Veikkolainen, V.; Ali, N.; Doroszko, M.; Kiviniemi, A.; Miinalainen, I.; Ohlsson, C.; Poutanen, M.; Rahman, N.; Elenius, K.; Vainio, S.J.; et al. Erbb4 regulates the oocyte microenvironment during folliculogenesis. Hum. Mol. Genet. 2020, 29, 2813–2830. [Google Scholar] [CrossRef]
  78. Clark, K.L.; George, J.W.; Przygrodzka, E.; Plewes, M.R.; Hua, G.; Wang, C.; Davis, J.S. Hippo Signaling in the Ovary: Emerging Roles in Development, Fertility, and Disease. Endocr. Rev. 2022, 43, 1074–1096. [Google Scholar] [CrossRef]
  79. Wang, C.; Jeong, K.; Jiang, H.; Guo, W.; Gu, C.; Lu, Y.; Liang, J. YAP/TAZ regulates the insulin signaling via IRS1/2 in endometrial cancer. Am. J. Cancer Res. 2016, 6, 996–1010. [Google Scholar]
  80. Tyrmi, J.S.; Arffman, R.K.; Pujol-Gualdo, N.; Kurra, V.; Morin-Papunen, L.; Sliz, E.; Piltonen, T.T.; Laisk, T.; Kettunen, J.; Laivuori, H. Leveraging Northern European population history: Novel low-frequency variants for polycystic ovary syndrome. Hum. Reprod. 2021, 37, 352–365. [Google Scholar] [CrossRef]
  81. Ruth, K.S.; Day, F.R.; Hussain, J.; Martínez-Marchal, A.; Aiken, C.E.; Azad, A.; Thompson, D.J.; Knoblochova, L.; Abe, H.; Tarry-Adkins, J.L.; et al. Genetic insights into biological mechanisms governing human ovarian ageing. Nature 2021, 596, 393–397. [Google Scholar] [CrossRef]
  82. Forslund, M.; Landin-Wilhelmsen, K.; Schmidt, J.; Brännström, M.; Trimpou, P.; Dahlgren, E. Higher menopausal age but no differences in parity in women with polycystic ovary syndrome compared with controls. Acta Obstet. Gynecol. Scand. 2018, 98, 320–326. [Google Scholar] [CrossRef]
  83. de Ziegler, D.; Pirtea, P.; Fanchin, R.; Ayoubi, J.M. Ovarian reserve in polycystic ovary syndrome: More, but for how long? Fertil. Steril. 2018, 109, 448–449. [Google Scholar] [CrossRef] [PubMed]
  84. Al-Awadi, A.M.; Sarray, S.; Arekat, M.R.; Saleh, L.R.; Mahmood, N.; Almawi, W.Y. The high-molecular weight multimer form of adiponectin is a useful marker of polycystic ovary syndrome in Bahraini Arab women. Clin. Nutr. ESPEN 2016, 13, e33–e38. [Google Scholar] [CrossRef]
  85. Yang, Z.; Yang, X.; Xu, J.; Sun, Y.; Shi, Y.; Fang, S. Association between Adiponectin Receptor 1 Gene Polymorphism and Insulin Resistance in Chinese Patients with Polycystic Ovary Syndrome. Gynecol. Obstet. Investig. 2013, 77, 45–49. [Google Scholar] [CrossRef] [PubMed]
  86. Al-Awadi, A.M.; Babi, A.; Finan, R.R.; Atageldiyeva, K.; Shaimardanova, M.; Mustafa, F.E.; Mahmood, N.A.; Aimagambetova, G.; Almawi, W.Y. ADIPOQ gene polymorphisms and haplotypes linked to altered susceptibility to PCOS: A case–control study. Reprod. Biomed. Online 2022, 45, 995–1005. [Google Scholar] [CrossRef] [PubMed]
  87. Ezzidi, I.; Mtiraoui, N.; Ali, M.E.M.; Al Masoudi, A.; Abu Duhier, F. Adiponectin (ADIPOQ) gene variants and haplotypes in Saudi Arabian women with polycystic ovary syndrome (PCOS): A case–control study. Gynecol. Endocrinol. 2019, 36, 66–71. [Google Scholar] [CrossRef]
  88. A Alfaqih, M.; Khader, Y.S.; Al-Dwairi, A.N.; Alzoubi, A.; Othman, A.-S.; Hatim, A. Lower Levels of Serum Adiponectin and the T Allele of rs1501299 of the ADIPOQ Gene Are Protective against Polycystic Ovarian Syndrome in Jordan. Korean J. Fam. Med. 2018, 39, 108–113. [Google Scholar] [CrossRef]
  89. Singh, A.; Bora, P.; Krishna, A. Direct action of adiponectin ameliorates increased androgen synthesis and reduces insulin receptor expression in the polycystic ovary. Biochem. Biophys. Res. Commun. 2017, 488, 509–515. [Google Scholar] [CrossRef]
  90. Zhang, F.; Lupski, J.R. Non-coding genetic variants in human disease: Figure 1. Hum. Mol. Genet. 2015, 24, R102–R110. [Google Scholar] [CrossRef]
  91. Zhu, Y.; Tazearslan, C.; Suh, Y. Challenges and progress in interpretation of non-coding genetic variants associated with human disease. Exp. Biol. Med. 2017, 242, 1325–1334. [Google Scholar] [CrossRef] [PubMed]
  92. Fitipaldi, H.; Franks, P.W. Ethnic, gender and other sociodemographic biases in genome-wide association studies for the most burdensome non-communicable diseases: 2005–2022. Hum. Mol. Genet. 2022, 32, 520–532. [Google Scholar] [CrossRef]
  93. Fatumo, S.; Chikowore, T.; Choudhury, A.; Ayub, M.; Martin, A.R.; Kuchenbaecker, K. A roadmap to increase diversity in genomic studies. Nat. Med. 2022, 28, 243–250. [Google Scholar] [CrossRef]
  94. Peterson, R.E.; Kuchenbaecker, K.; Walters, R.K.; Chen, C.-Y.; Popejoy, A.B.; Periyasamy, S.; Lam, M.; Iyegbe, C.; Strawbridge, R.J.; Brick, L.; et al. Genome-wide Association Studies in Ancestrally Diverse Populations: Opportunities, Methods, Pitfalls, and Recommendations. Cell 2019, 179, 589–603. [Google Scholar] [CrossRef]
  95. Zhang, Y.; Movva, V.C.; Williams, M.S.; Lee, M.T.M. Polycystic Ovary Syndrome Susceptibility Loci Inform Disease Etiological Heterogeneity. J. Clin. Med. 2021, 10, 2688. [Google Scholar] [CrossRef]
  96. Liu, P.; Huang, L.; Song, C.-Q. Editorial: Genome editing applications of CRISPR/Cas9 in metabolic diseases, hormonal system and cancer research. Front. Endocrinol. 2023, 14, 1256966. [Google Scholar] [CrossRef]
  97. Lidaka, L.; Bekere, L.; Lazdane, G.; Lazovska, M.; Dzivite-Krisane, I.; Gailite, L. Role of Single Nucleotide Variants in the YAP1 Gene in Adolescents with Polycystic Ovary Syndrome. Biomedicines 2022, 10, 1688. [Google Scholar] [CrossRef]
  98. Padder, K.A.; Yousuf, M.A.; Jahan, N.; Yousuf, S.D.; Ganie, M.A. Interplay between elevated RAB5B gene expression and insulin resistance among women with PCOS-insights from a case-control study. Endocrine 2024, 88, 323–329. [Google Scholar] [CrossRef]
  99. Hossain, M.A. CRISPR-Cas9: A fascinating journey from bacterial immune system to human gene editing. Prog. Mol. Biol. Transl. Sci. 2021, 178, 63–83. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, H.; La Russa, M.; Qi, L.S. CRISPR/Cas9 in Genome Editing and Beyond. Annu. Rev. Biochem. 2016, 85, 227–264. [Google Scholar] [CrossRef] [PubMed]
  101. Khan, F.J.; Yuen, G.; Luo, J. Multiplexed CRISPR/Cas9 gene knockout with simple crRNA:tracrRNA co-transfection. Cell Biosci. 2019, 9, 41. [Google Scholar] [CrossRef]
  102. Kantor, A.; McClements, M.E.; MacLaren, R.E. CRISPR-Cas9 DNA Base-Editing and Prime-Editing. Int. J. Mol. Sci. 2020, 21, 6240. [Google Scholar] [CrossRef]
  103. Wang, S.-W.; Gao, C.; Zheng, Y.-M.; Yi, L.; Lu, J.-C.; Huang, X.-Y.; Cai, J.-B.; Zhang, P.-F.; Cui, Y.-H.; Ke, A.-W. Current applications and future perspective of CRISPR/Cas9 gene editing in cancer. Mol. Cancer 2022, 21, 57. [Google Scholar] [CrossRef]
  104. Reh, W.A.; Vasquez, K.M. Gene Targeting by Homologous Recombination. In eLS; Available online: https://onlinelibrary.wiley.com/doi/abs/10.1002/9780470015902.a0005988.pub2 (accessed on 26 October 2025).
  105. Gupta, D.; Bhattacharjee, O.; Mandal, D.; Sen, M.K.; Dey, D.; Dasgupta, A.; Kazi, T.A.; Gupta, R.; Sinharoy, S.; Acharya, K.; et al. CRISPR-Cas9 system: A new-fangled dawn in gene editing. Life Sci. 2019, 232, 116636. [Google Scholar] [CrossRef]
  106. Li, H.; Yang, Y.; Hong, W.; Huang, M.; Wu, M.; Zhao, X. Applications of genome editing technology in the targeted therapy of human diseases: Mechanisms, advances and prospects. Signal Transduct. Target. Ther. 2020, 5, 1. [Google Scholar] [CrossRef] [PubMed]
  107. Sahin, G.N.; Seli, E. Gene editing using CRISPR-Cas9 technology: Potential implications in assisted reproduction. Curr. Opin. Obstet. Gynecol. 2025, 37, 141–148. [Google Scholar] [CrossRef]
  108. Guller, A.S.; Kayabolen, G.N.S.; Soyler, G.; Comar, M.Y.; Duzgun, I.Y.; Sivaslioglu, A.; Karahuseyinoglu, S. O-104 The Impact of application of CRISPR /dCas9 systems for increasing the expression of FSH receptor in human granulosa cells. Hum. Reprod. 2023, 38, dead093.127. [Google Scholar] [CrossRef]
  109. Livadas, S.; Paparodis, R.; Anagnostis, P.; Gambineri, A.; Bjekić-Macut, J.; Petrović, T.; Yildiz, B.O.; Micić, D.; Mastorakos, G.; Macut, D. Assessment of Type 2 Diabetes Risk in Young Women with Polycystic Ovary Syndrome. Diagnostics 2023, 13, 2067. [Google Scholar] [CrossRef]
  110. Srivastav, A.K.; Mishra, M.K.; Lillard, J.W.; Singh, R. Transforming Pharmacogenomics and CRISPR Gene Editing with the Power of Artificial Intelligence for Precision Medicine. Pharmaceutics 2025, 17, 555. [Google Scholar] [CrossRef] [PubMed]
  111. Matboli, M.; Kamel, M.M.; Essawy, N.; Bekhit, M.M.; Abdulrahman, B.; Mohamed, G.F.; Eissa, S. Identification of Novel Insulin Resistance Related ceRNA Network in T2DM and Its Potential Editing by CRISPR/Cas9. Int. J. Mol. Sci. 2021, 22, 8129. [Google Scholar] [CrossRef] [PubMed]
  112. Magoffin, D.A. Ovarian theca cell. Int. J. Biochem. Cell Biol. 2005, 37, 1344–1349. [Google Scholar] [CrossRef]
  113. Richards, J.S.; A Ren, Y.; Candelaria, N.; E Adams, J.; Rajkovic, A. Ovarian Follicular Theca Cell Recruitment, Differentiation, and Impact on Fertility: 2017 Update. Endocr. Rev. 2017, 39, 1–20. [Google Scholar] [CrossRef]
  114. Dong, J.; Rees, D.A. Polycystic ovary syndrome: Pathophysiology and therapeutic opportunities. BMJ Med. 2023, 2, e000548. [Google Scholar] [CrossRef] [PubMed]
  115. Ma, Y.; Andrisse, S.; Chen, Y.; Childress, S.; Xue, P.; Wang, Z.; Jones, D.; Ko, C.; Divall, S.; Wu, S. Androgen Receptor in the Ovary Theca Cells Plays a Critical Role in Androgen-Induced Reproductive Dysfunction. Endocrinology 2016, 158, 98–108. [Google Scholar] [CrossRef]
  116. Liu, Y.; Zhu, J.; Yang, Y.; Chen, Z.; Zhou, Y.; Fei, W.; Zhang, X.; Zheng, Y. Extracellular matrix dysregulation in PCOS: Pathogenesis, therapeutic strategies, and innovative technologies. J. Biol. Eng. 2025, 19, 61. [Google Scholar] [CrossRef]
  117. Waterbury, J.S.; E Teves, M.; Gaynor, A.; Han, A.X.; Mavodza, G.; Newell, J.; Strauss, J.F.; McAllister, J.M. The PCOS GWAS Candidate Gene ZNF217 Influences Theca Cell Expression of DENND1A.V2, CYP17A1, and Androgen Production. J. Endocr. Soc. 2022, 6, bvac078. [Google Scholar] [CrossRef]
  118. Teves, M.E.; Modi, B.P.; Kulkarni, R.; Han, A.X.; Marks, J.S.; Subler, M.A.; Windle, J.; Newall, J.M.; McAllister, J.M.; Strauss, J.F., III. Human DENND1A.V2 Drives Cyp17a1 Expression and Androgen Production in Mouse Ovaries and Adrenals. Int. J. Mol. Sci. 2020, 21, 2545. [Google Scholar] [CrossRef]
  119. Joseph, S.; Ubba, V.; Wang, Z.; Feng, M.; Dsilva, M.K.; Suero, S.; Waheed, D.; Snyder, N.W.; Yang, X.; Wang, H.; et al. Ovarian-Specific Cyp17A1 Overexpression in Female Mice: A Novel Model of Endogenous Testosterone Excess. Endocrinology 2025, 166, bqaf071. [Google Scholar] [CrossRef] [PubMed]
  120. Udhane, S.S.; Flück, C.E. Regulation of human (adrenal) androgen biosynthesis—New insights from novel throughput technology studies. Biochem. Pharmacol. 2016, 102, 20–33. [Google Scholar] [CrossRef]
  121. Lee, J.; Yamazaki, T.; Dong, H.; Jefcoate, C. A single cell level measurement of StAR expression and activity in adrenal cells. Mol. Cell. Endocrinol. 2017, 441, 22–30. [Google Scholar] [CrossRef] [PubMed]
  122. Wickenheisser, J.K.; Biegler, J.M.; Nelson-DeGrave, V.L.; Legro, R.S.; Strauss, J.F.; McAllister, J.M. Cholesterol Side-Chain Cleavage Gene Expression in Theca Cells: Augmented Transcriptional Regulation and mRNA Stability in Polycystic Ovary Syndrome. PLoS ONE 2012, 7, e48963. [Google Scholar] [CrossRef]
  123. Owens, L.A.; Kristensen, S.G.; Lerner, A.; Christopoulos, G.; Lavery, S.; Hanyaloglu, A.C.; Hardy, K.; Andersen, C.Y.; Franks, S. Gene Expression in Granulosa Cells From Small Antral Follicles From Women With or Without Polycystic Ovaries. J. Clin. Endocrinol. Metab. 2019, 104, 6182–6192. [Google Scholar] [CrossRef]
  124. Dumesic, D.A.; Richards, J.S. Ontogeny of the ovary in polycystic ovary syndrome. Fertil. Steril. 2013, 100, 23–38. [Google Scholar] [CrossRef]
  125. Kong, F.-S.; Feng, J.; Yao, J.-P.; Lu, Y.; Guo, T.; Sun, M.; Ren, C.-Y.; Jin, Y.-Y.; Ma, Y.; Chen, J.-H. Dysregulated RNA editing of EIF2AK2 in polycystic ovary syndrome: Clinical relevance and functional implications. BMC Med. 2024, 22, 229. [Google Scholar] [CrossRef]
  126. Kong, F.-S.; Lu, Z.; Zhou, Y.; Lu, Y.; Ren, C.-Y.; Jia, R.; Zeng, B.; Huang, P.; Wang, J.; Ma, Y.; et al. Transcriptome analysis identification of A-to-I RNA editing in granulosa cells associated with PCOS. Front. Endocrinol. 2023, 14, 1170957. [Google Scholar] [CrossRef] [PubMed]
  127. Tang, W.; Fei, Y.; Page, M. Biological Significance of RNA Editing in Cells. Mol. Biotechnol. 2012, 52, 91–100. [Google Scholar] [CrossRef] [PubMed]
  128. Barbarroja, N.; Rodriguez-Cuenca, S.; Nygren, H.; Camargo, A.; Pirraco, A.; Relat, J.; Cuadrado, I.; Pellegrinelli, V.; Medina-Gomez, G.; Lopez-Pedrera, C.; et al. Increased Dihydroceramide/Ceramide Ratio Mediated by Defective Expression of degs1 Impairs Adipocyte Differentiation and Function. Diabetes 2014, 64, 1180–1192. [Google Scholar] [CrossRef]
  129. Chappell, N.R.; Zhou, B.; Hosseinzadeh, P.; Schutt, A.; Gibbons, W.E.; Blesson, C.S. Hyperandrogenemia alters mitochondrial structure and function in the oocytes of obese mouse with polycystic ovary syndrome. F&S Sci. 2020, 2, 101–112. [Google Scholar] [CrossRef]
  130. Rudnicka, E.; Suchta, K.; Grymowicz, M.; Calik-Ksepka, A.; Smolarczyk, K.; Duszewska, A.M.; Smolarczyk, R.; Meczekalski, B. Chronic Low Grade Inflammation in Pathogenesis of PCOS. Int. J. Mol. Sci. 2021, 22, 3789. [Google Scholar] [CrossRef] [PubMed]
  131. Hesampour, F.; Jahromi, B.N.; Tahmasebi, F.; Gharesi-Fard, B. Association between Interleukin-32 and Interleukin-17A Single Nucleotide Polymorphisms and Serum Levels with Polycystic Ovary Syndrome. Iran. J. Allergy Asthma Immunol. 2019, 18, 91–99. [Google Scholar] [CrossRef]
  132. Gong, Y.; Luo, S.; Fan, P.; Zhu, H.; Li, Y.; Huang, W. Growth hormone activates PI3K/Akt signaling and inhibits ROS accumulation and apoptosis in granulosa cells of patients with polycystic ovary syndrome. Reprod. Biol. Endocrinol. 2020, 18, 121. [Google Scholar] [CrossRef]
  133. Wang, T.; Zhang, J.; Hu, M.; Zhang, Y.; Cui, P.; Li, X.; Li, J.; Vestin, E.; Brännström, M.; Shao, L.R.; et al. Differential Expression Patterns of Glycolytic Enzymes and Mitochondria-Dependent Apoptosis in PCOS Patients with Endometrial Hyperplasia, an Early Hallmark of Endometrial Cancer, In Vivo and the Impact of Metformin In Vitro. Int. J. Biol. Sci. 2019, 15, 714–725. [Google Scholar] [CrossRef]
  134. Yu, Y.; Li, G.; He, X.; Lin, Y.; Chen, Z.; Lin, X.; Xu, H. MicroRNA-21 regulate the cell apoptosis and cell proliferation of polycystic ovary syndrome (PCOS) granulosa cells through target toll like receptor TLR8. Bioengineered 2021, 12, 5789–5796. [Google Scholar] [CrossRef] [PubMed]
  135. Zhao, F.; Wu, L.; Wang, Q.; Zhao, X.; Chen, T.; Yin, C.; Yan, L.; Yang, X. Insulin-like growth factor 2 mRNA-binding protein 2-regulated alternative splicing of nuclear factor 1 C-type causes excessive granulosa cell proliferation in polycystic ovary syndrome. Cell Prolif. 2022, 55, e13216. [Google Scholar] [CrossRef]
  136. Li, R.Z.; Hou, J.; Wei, Y.; Luo, X.; Ye, Y.; Zhang, Y. hnRNPDL extensively regulates transcription and alternative splicing. Gene 2019, 687, 125–134. [Google Scholar] [CrossRef]
  137. Berardo, A.; Lornage, X.; Johari, M.; Evangelista, T.; Cejas, C.; Barroso, F.; Dubrovsky, A.; Bui, M.T.; Brochier, G.; Saccoliti, M.; et al. HNRNPDL-related muscular dystrophy: Expanding the clinical, morphological and MRI phenotypes. J. Neurol. 2019, 266, 2524–2534. [Google Scholar] [CrossRef] [PubMed]
  138. Nishikura, K. Functions and Regulation of RNA Editing by ADAR Deaminases. Annu. Rev. Biochem. 2010, 79, 321–349. [Google Scholar] [CrossRef]
  139. Meyer, C.; Garzia, A.; Mazzola, M.; Gerstberger, S.; Molina, H.; Tuschl, T. The TIA1 RNA-Binding Protein Family Regulates EIF2AK2-Mediated Stress Response and Cell Cycle Progression. Mol. Cell 2018, 69, 622–635.e6. [Google Scholar] [CrossRef]
  140. Cesaro, T.; Hayashi, Y.; Borghese, F.; Vertommen, D.; Wavreil, F.; Michiels, T. PKR activity modulation by phosphomimetic mutations of serine residues located three aminoacids upstream of double-stranded RNA binding motifs. Sci. Rep. 2021, 11, 9188. [Google Scholar] [CrossRef] [PubMed]
  141. Li, Y.; Wu, H.; Guo, Y.; Wei, C.; Guan, L.; Ju, W.; Lian, F. Cangfu Daotan Wan alleviates polycystic ovary syndrome with phlegm-dampness syndrome via disruption of the PKP3/ERCC1/MAPK axis. J. Ovarian Res. 2023, 16, 134. [Google Scholar] [CrossRef]
  142. Das, S.; Maizels, E.T.; DeManno, D.; Clair, E.S.; A Adam, S.; Hunzicker-Dunn, M. A stimulatory role of cyclic adenosine 3′,5′-monophosphate in follicle-stimulating hormone-activated mitogen-activated protein kinase signaling pathway in rat ovarian granulosa cells. Endocrinology 1996, 137, 967–974. [Google Scholar] [CrossRef]
  143. Tajima, K.; Dantes, A.; Yao, Z.; Sorokina, K.; Kotsuji, F.; Seger, R.; Amsterdam, A. Down-Regulation of Steroidogenic Response to Gonadotropins in Human and Rat Preovulatory Granulosa Cells Involves Mitogen-Activated Protein Kinase Activation and Modulation of DAX-1 and Steroidogenic Factor-1. J. Clin. Endocrinol. Metab. 2003, 88, 2288–2299. [Google Scholar] [CrossRef]
  144. Gruijters, M.J.; Visser, J.A.; Durlinger, A.L.; Themmen, A.P. Anti-Müllerian hormone and its role in ovarian function. Mol. Cell. Endocrinol. 2003, 211, 85–90. [Google Scholar] [CrossRef]
  145. Ke, Y.; Tang, D.; Yang, Q.; Zhao, H.; Zheng, J.; Zhu, C. Anti-Müllerian hormone in PCOS: Molecular regulation and emerging therapeutic strategies. Biomol. Biomed. 2025, 26, 173–185. [Google Scholar] [CrossRef] [PubMed]
  146. Bouchard, M.F.; Bergeron, F.; Delaney, J.G.; Harvey, L.-M.; Viger, R.S. In Vivo Ablation of the Conserved GATA-Binding Motif in the Amh Promoter Impairs Amh Expression in the Male Mouse. Endocrinology 2019, 160, 817–826. [Google Scholar] [CrossRef] [PubMed]
  147. Manuylov, N.; Zhou, B.; Ma, Q.; Fox, S.; Pu, W.; Tevosian, S. Conditional ablation of Gata4 and Fog2 genes in mice reveals their distinct roles in mammalian sexual differentiation. Dev. Biol. 2011, 353, 229–241. [Google Scholar] [CrossRef]
  148. Liu, Z.; Dai, L.; Sun, T.; Liu, Y.; Bao, Y.; Gu, M.; Fu, S.; He, X.; Shi, C.; Wang, Y.; et al. Massively Parallel CRISPR-Cas9 Knockout Screening in Sheep Granulosa Cells for FSH Response Genes. Animals 2024, 14, 898. [Google Scholar] [CrossRef]
  149. Kumar, S.; Punetha, M.; Jose, B.; Bharati, J.; Khanna, S.; Sonwane, A.; Green, J.A.; Whitworth, K.; Sarkar, M. Modulation of granulosa cell function via CRISPR-Cas fuelled editing of BMPR-IB gene in goats (Capra hircus). Sci. Rep. 2020, 10, 20446. [Google Scholar] [CrossRef]
  150. Oktem, O.; Urman, B. Understanding follicle growth in vivo. Hum. Reprod. 2010, 25, 2944–2954. [Google Scholar] [CrossRef]
  151. Takov, E.C.V. Embryology, Ovarian Follicle Development. In StatPearls [Internet]; StatPearls Publishing: St. Petersburg, FL, USA, 2025. [Google Scholar]
  152. Woodruff, T.K.; Shea, L.D. The Role of the Extracellular Matrix in Ovarian Follicle Development. Reprod. Sci. 2007, 14, 6–10. [Google Scholar] [CrossRef] [PubMed]
  153. Franks, S.; Stark, J.; Hardy, K. Follicle dynamics and anovulation in polycystic ovary syndrome. Hum. Reprod. Updat. 2008, 14, 367–378. [Google Scholar] [CrossRef]
  154. Wang, K.; Li, Y. Signaling pathways and targeted therapeutic strategies for polycystic ovary syndrome. Front. Endocrinol. 2023, 14, 1191759. [Google Scholar] [CrossRef]
  155. Ruan, Y.; Li, X.; Wang, X.; Zhai, G.; Lou, Q.; Jin, X.; He, J.; Mei, J.; Xiao, W.; Gui, J.; et al. New insights into the all-testis differentiation in zebrafish with compromised endogenous androgen and estrogen synthesis. PLoS Genet. 2024, 20, e1011170. [Google Scholar] [CrossRef]
  156. Lau, E.S.-W.; Zhang, Z.; Qin, M.; Ge, W. Knockout of Zebrafish Ovarian Aromatase Gene (cyp19a1a) by TALEN and CRISPR/Cas9 Leads to All-male Offspring Due to Failed Ovarian Differentiation. Sci. Rep. 2016, 6, 37357. [Google Scholar] [CrossRef]
  157. Carver, J.J.; Amato, C.M.; Yao, H.H.-C.; Zhu, Y. Adamts9 is required for the development of primary ovarian follicles and maintenance of female sex in zebrafish. Biol. Reprod. 2024, 111, 1107–1128. [Google Scholar] [CrossRef] [PubMed]
  158. Kushawaha, B.; Pelosi, E. Spotlight on Proteases: Roles in Ovarian Health and Disease. Cells 2025, 14, 921. [Google Scholar] [CrossRef] [PubMed]
  159. Wu, K.; Yue, Y.; Zhou, L.; Zhang, Z.; Shan, H.; He, H.; Ge, W. Disrupting Amh and androgen signaling reveals their distinct roles in zebrafish gonadal differentiation and gametogenesis. Commun. Biol. 2025, 8, 371. [Google Scholar] [CrossRef]
  160. Zhang, Z.; Zhu, B.; Chen, W.; Ge, W. Anti-Müllerian hormone (Amh/amh) plays dual roles in maintaining gonadal homeostasis and gametogenesis in zebrafish. Mol. Cell. Endocrinol. 2020, 517, 110963. [Google Scholar] [CrossRef]
  161. Chen, Z.; Liu, L.; Xi, X.; Burn, M.; Karakaya, C.; Kallen, A.N. Aberrant H19 Expression Disrupts Ovarian Cyp17 and Testosterone Production and Is Associated with Polycystic Ovary Syndrome in Women. Reprod. Sci. 2021, 29, 1357–1367. [Google Scholar] [CrossRef] [PubMed]
  162. Chen, Z.; Yu, H.; Shi, X.; Warren, C.R.; Lotta, L.A.; Friesen, M.; Meissner, T.B.; Langenberg, C.; Wabitsch, M.; Wareham, N.; et al. Functional Screening of Candidate Causal Genes for Insulin Resistance in Human Preadipocytes and Adipocytes. Circ. Res. 2019, 126, 330–346. [Google Scholar] [CrossRef]
  163. Kahn, C.R. Knockout Mice Challenge our Concepts of Glucose Homeostasis and the Pathogenesis of Diabetes. J. Diabetes Res. 2003, 4, 169–182. [Google Scholar] [CrossRef]
  164. Garofalo, R.S.; Orena, S.J.; Rafidi, K.; Torchia, A.J.; Stock, J.L.; Hildebrandt, A.L.; Coskran, T.; Black, S.C.; Brees, D.J.; Wicks, J.R.; et al. Severe diabetes, age-dependent loss of adipose tissue, and mild growth deficiency in mice lacking Akt2/PKBβ. J. Clin. Investig. 2003, 112, 197–208. [Google Scholar] [CrossRef] [PubMed]
  165. Miki, H.; Yamauchi, T.; Suzuki, R.; Komeda, K.; Tsuchida, A.; Kubota, N.; Terauchi, Y.; Kamon, J.; Kaburagi, Y.; Matsui, J.; et al. Essential Role of Insulin Receptor Substrate 1 (IRS-1) and IRS-2 in Adipocyte Differentiation. Mol. Cell. Biol. 2001, 21, 2521–2532. [Google Scholar] [CrossRef]
  166. Kamble, P.G.; Hetty, S.; Vranic, M.; Almby, K.; Castillejo-López, C.; Abalo, X.M.; Pereira, M.J.; Eriksson, J.W. Proof-of-concept for CRISPR/Cas9 gene editing in human preadipocytes: Deletion of FKBP5 and PPARG and effects on adipocyte differentiation and metabolism. Sci. Rep. 2020, 10, 10565. [Google Scholar] [CrossRef]
  167. Claussnitzer, M.; Dankel, S.N.; Kim, K.-H.; Quon, G.; Meuleman, W.; Haugen, C.; Glunk, V.; Sousa, I.S.; Beaudry, J.L.; Puviindran, V.; et al. FTO Obesity Variant Circuitry and Adipocyte Browning in Humans. N. Engl. J. Med. 2015, 373, 895–907. [Google Scholar] [CrossRef]
  168. Roh, J.-I.; Lee, J.; Park, S.U.; Kang, Y.-S.; Lee, J.; Oh, A.-R.; Choi, D.J.; Cha, J.-Y.; Lee, H.-W. CRISPR-Cas9-mediated generation of obese and diabetic mouse models. Exp. Anim. 2018, 67, 229–237. [Google Scholar] [CrossRef] [PubMed]
  169. Bao, D.; Ma, Y.; Zhang, X.; Guan, F.; Chen, W.; Gao, K.; Qin, C.; Zhang, L. Preliminary Characterization of a Leptin Receptor Knockout Rat Created by CRISPR/Cas9 System. Sci. Rep. 2015, 5, 15942. [Google Scholar] [CrossRef]
  170. Hao, H.; Lin, R.; Li, Z.; Shi, W.; Huang, T.; Niu, J.; Han, J.; Li, Q. MC4R deficiency in pigs results in hyperphagia and ultimately hepatic steatosis without high-fat diet. Biochem. Biophys. Res. Commun. 2019, 520, 651–656. [Google Scholar] [CrossRef] [PubMed]
  171. Uhlenhaut, N.H.; Jakob, S.; Anlag, K.; Eisenberger, T.; Sekido, R.; Kress, J.; Treier, A.-C.; Klugmann, C.; Klasen, C.; Holter, N.I.; et al. Somatic Sex Reprogramming of Adult Ovaries to Testes by FOXL2 Ablation. Cell 2009, 139, 1130–1142. [Google Scholar] [CrossRef]
  172. Wei, C.; Wang, F.; Liu, W.; Zhao, W.; Yang, Y.; Li, K.; Xiao, L.; Shen, J. CRISPR/Cas9 targeting of the androgen receptor suppresses the growth of LNCaP human prostate cancer cells. Mol. Med. Rep. 2017, 17, 2901–2906. [Google Scholar] [CrossRef]
  173. Lin, Z.; Li, Y.; Xu, Y.; Wang, Q.; Namgoong, S.; Cui, X.; Kim, N. Effects of Growth Differentiation Factor 9 and Bone Morphogenetic Protein 15 on the in vitro Maturation of Porcine Oocytes. Reprod. Domest. Anim. 2013, 49, 219–227. [Google Scholar] [CrossRef]
  174. Fountas, S.; Petinaki, E.; Bolaris, S.; Kargakou, M.; Dafopoulos, S.; Zikopoulos, A.; Moustakli, E.; Sotiriou, S.; Dafopoulos, K. The Roles of GDF-9, BMP-15, BMP-4 and EMMPRIN in Folliculogenesis and In Vitro Fertilization. J. Clin. Med. 2024, 13, 3775. [Google Scholar] [CrossRef] [PubMed]
  175. McCarty, N.S.; Graham, A.E.; Studená, L.; Ledesma-Amaro, R. Multiplexed CRISPR technologies for gene editing and transcriptional regulation. Nat. Commun. 2020, 11, 1281. [Google Scholar] [CrossRef]
  176. Singh, S.; Pal, N.; Shubham, S.; Sarma, D.K.; Verma, V.; Marotta, F.; Kumar, M. Polycystic Ovary Syndrome: Etiology, Current Management, and Future Therapeutics. J. Clin. Med. 2023, 12, 1454. [Google Scholar] [CrossRef]
  177. Shi, J.; Gao, Q.; Cao, Y.; Fu, J. Dennd1a, a susceptibility gene for polycystic ovary syndrome, is essential for mouse embryogenesis. Dev. Dyn. 2019, 248, 351–362. [Google Scholar] [CrossRef]
  178. Shaaban, Z.; Khoradmehr, A.; Amiri-Yekta, A.; Nowzari, F.; Shirazi, M.R.J.; Tamadon, A. Pathophysiologic Mechanisms of Insulin Secretion and Signaling-Related Genes in Etiology of Polycystic Ovary Syndrome. Genet. Res. 2021, 2021, 7781823. [Google Scholar] [CrossRef]
  179. Yan, Y.; Desvignes, T.; Bremiller, R.; Wilson, C.; Dillon, D.; High, S.; Draper, B.; Buck, C.L.; Postlethwait, J. Gonadal soma controls ovarian follicle proliferation through Gsdf in zebrafish. Dev. Dyn. 2017, 246, 925–945. [Google Scholar] [CrossRef]
  180. Xu, J.; Dun, J.; Yang, J.; Zhang, J.; Lin, Q.; Huang, M.; Ji, F.; Huang, L.; You, X.; Lin, Y. Letrozole Rat Model Mimics Human Polycystic Ovarian Syndrome and Changes in Insulin Signal Pathways. Med. Sci. Monit. 2020, 26, e923073. [Google Scholar] [CrossRef] [PubMed]
  181. Poojary, P.S.; Nayak, G.; Panchanan, G.; Rao, A.; Das Kundapur, S.; Kalthur, S.G.; Mutalik, S.; Adiga, S.K.; Zhao, Y.; Bakkum-Gamez, J.; et al. Distinctions in PCOS Induced by Letrozole Vs Dehydroepiandrosterone with High-fat Diet in Mouse Model. Endocrinology 2022, 163, bqac097. [Google Scholar] [CrossRef] [PubMed]
  182. Uitterlinden, A.G. An Introduction to Genome-Wide Association Studies: GWAS for Dummies. Semin. Reprod. Med. 2016, 34, 196–204. [Google Scholar] [CrossRef]
  183. Tan, M.; Cheng, Y.; Zhong, X.; Yang, D.; Jiang, S.; Ye, Y.; Ding, M.; Guan, G.; Yang, D.; Zhao, X. LNK promotes granulosa cell apoptosis in PCOS via negatively regulating insulin-stimulated AKT-FOXO3 pathway. Aging 2021, 13, 4617–4633. [Google Scholar] [CrossRef]
  184. Han, S.; Zhang, Y.; Zheng, Y.; Liu, C.; Jiang, Y.; Zhao, S.; Zhao, H. Thada Is Dispensable for Female Fertility in Mice. Front. Endocrinol. 2022, 13, 787733. [Google Scholar] [CrossRef]
  185. Velmurugan, S.; Pauline, R.; Subbaraj, G.K. Association of candidate gene (INSR & THADA) polymorphism with polycystic ovary syndrome: Meta-analysis and statistical power analysis. J. Turk. Gynecol. Assoc. 2024, 25, 167–178. [Google Scholar] [CrossRef]
  186. Lit, K.K.; Zhirenova, Z.; Blocki, A. Insulin-like growth factor-binding protein 7 (IGFBP7): A microenvironment-dependent regulator of angiogenesis and vascular remodeling. Front. Cell Dev. Biol. 2024, 12, 1421438. [Google Scholar] [CrossRef] [PubMed]
  187. Chen, L.; Hui, L.; Shi, Y.; Wang, A.; Zhang, Y.; Yao, X.; Li, J. The role of IGFBP7 in the immune regulation of PCOS-like symptoms based on spleen transcriptome and TCR β CDR3 repertoire analysis. Mol. Cell. Endocrinol. 2025, 608, 112639. [Google Scholar] [CrossRef]
  188. Kosova, G.; Urbanek, M. Genetics of the polycystic ovary syndrome. Mol. Cell. Endocrinol. 2013, 373, 29–38. [Google Scholar] [CrossRef] [PubMed]
  189. Zou, J.; Li, Y.; Liao, N.; Liu, J.; Zhang, Q.; Luo, M.; Xiao, J.; Chen, Y.; Wang, M.; Chen, K.; et al. Identification of key genes associated with polycystic ovary syndrome (PCOS) and ovarian cancer using an integrated bioinformatics analysis. J. Ovarian Res. 2022, 15, 30. [Google Scholar] [CrossRef] [PubMed]
  190. Uddin, F.; Rudin, C.M.; Sen, T. CRISPR Gene Therapy: Applications, Limitations, and Implications for the Future. Front. Oncol. 2020, 10, 1387. [Google Scholar] [CrossRef]
  191. Cao, C.; Ma, Q.; Mo, S.; Shu, G.; Liu, Q.; Ye, J.; Gui, Y. Single-Cell RNA Sequencing Defines the Regulation of Spermatogenesis by Sertoli-Cell Androgen Signaling. Front. Cell Dev. Biol. 2021, 9. [Google Scholar] [CrossRef]
  192. Sun, Y.; Miao, N.; Sun, T. Detect accessible chromatin using ATAC-sequencing, from principle to applications. Hereditas 2019, 156, 29. [Google Scholar] [CrossRef]
  193. Sankaranarayanan, L.; Brewer, K.J.; Johnson, G.D.; Barrera, A.; Venukuttan, R.; Sisk, R.; Dunaif, A.; Reddy, T.E. Gene regulatory activity associated with PCOS revealed DENND1A-dependent testosterone production. bioRxiv 2024. bioRxiv:2024.2005.2023.595551. [Google Scholar] [CrossRef]
  194. Tee, M.K.; Speek, M.; Legeza, B.; Modi, B.; Teves, M.E.; McAllister, J.M.; Strauss, J.F.; Miller, W.L. Alternative splicing of DENND1A, a PCOS candidate gene, generates variant 2. Mol. Cell. Endocrinol. 2016, 434, 25–35. [Google Scholar] [CrossRef]
  195. McAllister, J.M.; Legro, R.S.; Modi, B.P.; Strauss, J.F. Functional genomics of PCOS: From GWAS to molecular mechanisms. Trends Endocrinol. Metab. 2015, 26, 118–124. [Google Scholar] [CrossRef] [PubMed]
  196. Gainder, S.; Sachdeva, G.; Suri, V.; Sachdeva, N.; Chopra, S. Comparison of the different PCOS phenotypes based on clinical metabolic, and hormonal profile, and their response to clomiphene. Indian J. Endocrinol. Metab. 2019, 23, 326–331. [Google Scholar] [CrossRef] [PubMed]
  197. Baba, T. Polycystic ovary syndrome: Criteria, phenotypes, race and ethnicity. Reprod. Med. Biol. 2025, 24, e12630. [Google Scholar] [CrossRef]
  198. Mózsik, L.; Hoekzema, M.; de Kok, N.A.W.; Bovenberg, R.A.L.; Nygård, Y.; Driessen, A.J.M. CRISPR-based transcriptional activation tool for silent genes in filamentous fungi. Sci. Rep. 2021, 11, 1118. [Google Scholar] [CrossRef]
  199. Makarova, K.S.; Grishin, N.V.; Shabalina, S.A.; Wolf, Y.I.; Koonin, E.V. A putative RNA-interference-based immune system in prokaryotes: Computational analysis of the predicted enzymatic machinery, functional analogies with eukaryotic RNAi, and hypothetical mechanisms of action. Biol. Direct 2006, 1, 7. [Google Scholar] [CrossRef]
  200. Kazemian, P.; Yu, S.-Y.; Thomson, S.B.; Birkenshaw, A.; Leavitt, B.R.; Ross, C.J.D. Lipid-Nanoparticle-Based Delivery of CRISPR/Cas9 Genome-Editing Components. Mol. Pharm. 2022, 19, 1669–1686. [Google Scholar] [CrossRef] [PubMed]
  201. Song, Z.; Tao, Y.; Liu, Y.; Li, J. Advances in delivery systems for CRISPR/Cas-mediated cancer treatment: A focus on viral vectors and extracellular vesicles. Front. Immunol. 2024, 15, 1444437. [Google Scholar] [CrossRef]
  202. Amarilla-Quintana, S.; Navarro, P.; Hernández, I.; Ramos, A.; Montero-Calle, A.; Cabezas-Sainz, P.; Barrero, M.J.; Megías, D.; Vilaplana-Martí, B.; Epifano, C.; et al. CRISPR targeting of FOXL2 c.402C>G mutation reduces malignant phenotype in granulosa tumor cells and identifies anti-tumoral compounds. Mol. Oncol. 2025, 19, 1092–1116. [Google Scholar] [CrossRef]
  203. Heinzelmann, E.; Piraino, F.; Costa, M.; Roch, A.; Norkin, M.; Garnier, V.; Homicsko, K.; Brandenberg, N. iPSC-derived and Patient-Derived Organoids: Applications and challenges in scalability and reproducibility as pre-clinical models. Curr. Res. Toxicol. 2024, 7, 100197. [Google Scholar] [CrossRef]
  204. Brooks, I.R.; Garrone, C.M.; Kerins, C.; Kiar, C.S.; Syntaka, S.; Xu, J.Z.; Spagnoli, F.M.; Watt, F.M. Functional genomics and the future of iPSCs in disease modeling. Stem Cell Rep. 2022, 17, 1033–1047. [Google Scholar] [CrossRef]
  205. Rubeis, G.; Steger, F. Risks and benefits of human germline genome editing: An ethical analysis. Asian Bioeth. Rev. 2018, 10, 133–141. [Google Scholar] [CrossRef]
  206. Chin, A.H.B.; Sun, N. Do not overlook the possibility of genome-edited somatic cells ending up in the human germline. J. Community Genet. 2024, 15, 749–752. [Google Scholar] [CrossRef]
  207. Kosicki, M.; Tomberg, K.; Bradley, A. Repair of double-strand breaks induced by CRISPR–Cas9 leads to large deletions and complex rearrangements. Nat. Biotechnol. 2018, 36, 765–771. [Google Scholar] [CrossRef]
  208. Aussel, C.; Cathomen, T.; Fuster-García, C. The hidden risks of CRISPR/Cas: Structural variations and genome integrity. Nat. Commun. 2025, 16, 7208. [Google Scholar] [CrossRef]
  209. Kleinstiver, B.P.; Pattanayak, V.; Prew, M.S.; Tsai, S.Q.; Nguyen, N.T.; Zheng, Z.; Joung, J.K. High-fidelity CRISPR–Cas9 nucleases with no detectable genome-wide off-target effects. Nature 2016, 529, 490–495. [Google Scholar] [CrossRef]
  210. Liu, W.; Li, L.; Jiang, J.; Wu, M.; Lin, P. Applications and challenges of CRISPR-Cas gene-editing to disease treatment in clinics. Precis. Clin. Med. 2021, 4, 179–191. [Google Scholar] [CrossRef] [PubMed]
  211. Guo, C.; Ma, X.; Gao, F.; Guo, Y. Off-target effects in CRISPR/Cas9 gene editing. Front. Bioeng. Biotechnol. 2023, 11, 1143157. [Google Scholar] [CrossRef] [PubMed]
  212. Kim, M.; Hwang, Y.; Lim, S.; Jang, H.-K.; Kim, H.-O. Advances in Nanoparticles as Non-Viral Vectors for Efficient Delivery of CRISPR/Cas9. Pharmaceutics 2024, 16, 1197. [Google Scholar] [CrossRef] [PubMed]
  213. Burdo, T.H.; Chen, C.; Kaminski, R.; Sariyer, I.K.; Mancuso, P.; Donadoni, M.; Smith, M.D.; Sariyer, R.; Caocci, M.; Liao, S.; et al. Preclinical safety and biodistribution of CRISPR targeting SIV in non-human primates. Gene Ther. 2023, 31, 224–233. [Google Scholar] [CrossRef] [PubMed]
  214. Palacios, A.M.; Korus, P.; Wilkens, B.G.C.; Heshmatpour, N.; Patnaik, S.R. Revolutionizing in vivo therapy with CRISPR/Cas genome editing: Breakthroughs, opportunities and challenges. Front. Genome Ed. 2024, 6, 1342193. [Google Scholar] [CrossRef]
  215. Pandey, V.; Sharma, S.; Pokharel, Y.R. Exploring CRISPR-Cas: The transformative impact of gene editing in molecular biology. Mol. Ther.-Nucleic Acids 2025, 36, 102717. [Google Scholar] [CrossRef]
  216. Zhuo, C.; Zhang, J.; Lee, J.-H.; Jiao, J.; Cheng, D.; Liu, L.; Kim, H.-W.; Tao, Y.; Li, M. Spatiotemporal control of CRISPR/Cas9 gene editing. Signal Transduct. Target. Ther. 2021, 6, 238. [Google Scholar] [CrossRef] [PubMed]
  217. Brokowski, C.; Adli, M. CRISPR Ethics: Moral Considerations for Applications of a Powerful Tool. J. Mol. Biol. 2019, 431, 88–101. [Google Scholar] [CrossRef] [PubMed]
  218. Ayanoğlu, F.B.; ELÇİN, A.E.; ELÇİN, Y.M. Bioethical issues in genome editing by CRISPR-Cas9 technology. Turk. J. Biol. 2020, 44, 110–120. [Google Scholar] [CrossRef]
Figure 1. Pathophysiology of PCOS: Reproductive-Metabolic Interactions. LH: Luteinizing hormone; FSH: Follicle-stimulating hormone; IR: Insulin resistance; SHBG: Sex-hormone binding globulin; IGFBP-1: Insulin-like growth factor-binding protein-1; IL-6: Interleukin 6; TNF-α: Tumor necrosis factor alpha. Arrows indicate the directional flow of hormonal, metabolic, and inflammatory signals between tissues, highlighting how hyperandrogenism, insulin resistance, and inflammation interact in a feedback cycle that sustains the reproductive and metabolic abnormalities observed in PCOS.
Figure 1. Pathophysiology of PCOS: Reproductive-Metabolic Interactions. LH: Luteinizing hormone; FSH: Follicle-stimulating hormone; IR: Insulin resistance; SHBG: Sex-hormone binding globulin; IGFBP-1: Insulin-like growth factor-binding protein-1; IL-6: Interleukin 6; TNF-α: Tumor necrosis factor alpha. Arrows indicate the directional flow of hormonal, metabolic, and inflammatory signals between tissues, highlighting how hyperandrogenism, insulin resistance, and inflammation interact in a feedback cycle that sustains the reproductive and metabolic abnormalities observed in PCOS.
Cells 14 01769 g001
Figure 2. CRISPR targeting of theca and granulosa cell genes in PCOS. LH: Luteinizing hormone; LHCGR: Luteinizing hormone/choriogonadotropin receptor; FSH: Follicle-stimulating hormone; FSHR: Follicle-stimulating hormone receptor; AMH: Anti-Müllerian hormone; AMHR2: Anti-Müllerian hormone type-2 receptor; KO: Knockout; OE: Overexpression. Arrows indicate the direction of hormonal signaling and gene regulation between theca and granulosa cells. Scissors icons represent CRISPR-mediated gene knockout (KO) or activation/overexpression (OE) used to model gene function in PCOS-related pathways.
Figure 2. CRISPR targeting of theca and granulosa cell genes in PCOS. LH: Luteinizing hormone; LHCGR: Luteinizing hormone/choriogonadotropin receptor; FSH: Follicle-stimulating hormone; FSHR: Follicle-stimulating hormone receptor; AMH: Anti-Müllerian hormone; AMHR2: Anti-Müllerian hormone type-2 receptor; KO: Knockout; OE: Overexpression. Arrows indicate the direction of hormonal signaling and gene regulation between theca and granulosa cells. Scissors icons represent CRISPR-mediated gene knockout (KO) or activation/overexpression (OE) used to model gene function in PCOS-related pathways.
Cells 14 01769 g002
Figure 3. CRISPR knockouts in metabolic pathways of PCOS. GLUT4: Glucose transporter type 4.
Figure 3. CRISPR knockouts in metabolic pathways of PCOS. GLUT4: Glucose transporter type 4.
Cells 14 01769 g003
Table 1. Genetic Landscape of PCOS and CRISPR-Relevant Targets.
Table 1. Genetic Landscape of PCOS and CRISPR-Relevant Targets.
Gene Role in PCOSPotential CRISPR Application
DENND1A [57,58]Overexpressed DENND1A isoform leads to excess androgen production Perform knockout to decrease androgen biosynthesis
THADA [59]Variants are linked to defects in energy metabolism Model metabolic dysfunction or correct variants to improve insulin sensitivity
LHCGR [53,63]Altered gene expression results in impaired folliculogenesisEdit variants to restore normal receptor function
FSHR [53,62,63]Reduced receptor sensitivity to FSH leads to defective follicle development and ovulatory dysfunctionCRISPRa to upregulate FSHR expression and increase responsiveness to FSH
INSR [60]Polymorphisms disrupt insulin receptor signaling, which contributes to IRTarget and excise specific INSR polymorphisms to restore normal receptor function in metabolic tissues
YAP1 [97]Increased activity in variants is associated with disruption in cellular processes and enlarged ovariesCRISPRi to downregulate YAP1 overexpression in ovarian granulosa cells
RAB5B [98]Upregulated expression is linked to IR Correct or knockout RAB5B to restore normal insulin signaling
HMGA2 [54]Altered expression is associated with hyperinsulinemia and IRModel HMGA2 to further understand metabolic effects
KISS1 [64,65]Polymorphisms exacerbate ovulatory dysfunction and contribute to abnormal BMI and LH levels Target SNPs to restore normal GnRH feedback and ovulation
LEPR [69] Variants disrupt leptin binding, impair insulin pathways, and increase IR Edit variants to improve insulin sensitivity and normalize leptin signaling
SOD2 [72,73]A16V variant could result in cellular damage and impair gonadotropin balanceTarget and edit variant to limit cellular injury and normalize hormone levels
ERBB4 [77]Dysfunction can lead to pronounced reproductive and metabolic abnormalities Create gene knockout models to further investigate how it induces PCOS features
WWTR1 [79]Altered expression can contribute to infertility, IR, and influences response to metformin Inhibit gene to enhance metformin efficacy
CHEK2 [81,82]Mutations in CHEK2 result in irregular oocyte apoptosis and follicular excess Develop models to better understand how variants contribute to PCOS
ADIPOQ [84,86]ADIPOQ polymorphisms are associated with reduced adiponectin levels Model ADIPOQ SNPs to study adiponectin signaling and ethnic-specific effects
ADIPOR1 [85,89]Variants impair adiponectin signaling and are linked to IRUse mouse models to test the effect of treatment on clinical characteristics
FSH: Follicle-stimulating hormone; CRISPRa: CRISPR activation; FSHR: Follicle-stimulating hormone receptor; IR: Insulin resistance; CRISPRi: CRISPR interference; BMI: Body mass index; LH: Luteinizing hormone; SNP: Single-nucleotide polymorphism; GnRH: Gonadotropin-releasing hormone.
Table 2. CRISPR Models Applied to PCOS-Relevant Genes.
Table 2. CRISPR Models Applied to PCOS-Relevant Genes.
Target GeneModel (Species/Cell)Key FindingsPCOS RelevanceModel Advantages and Limitations
FSHR [108]Human granulosa cellsdCas9 activation increased FSHR expression (strongest at 5 min with Gonal-f); estradiol levels also elevatedDemonstrates CRISPR modulation of gonadotropin signaling and steroidogenesisHuman-origin cell model; directly relevant to ovarian physiology. However, the study was conducted in a cell line because primary granulosa cells are difficult to target for epigenome editing.
IRS1 [162]Human adipocytesLoss of function led to insulin-stimulated AKT2 phosphorylation & glucose uptake; altered lipid metabolismLinks IRS1 loss to IR & metabolic features of PCOSAbundant source of adipogenic cells and is suitable for gene editing due to their proliferation ability. The knockout of the effector genes cannot fully replicate the regulatory state.
PPARG [166]Human preadipocytesKnockout of PPARG prevents differentiation of preadipocytes into adipocytesShows how PPARG plays a role in regulating adipogenesisExcluding selection markers enhanced knockout efficiency and allowed cells to preserve a high differentiation capacity, but inserting random indels may generate undesired epigenetic changes and result in false positives.
FTO (rs1421085) [167] CRISPR repair of the risk allele restored ARID5B binding, repressed IRX3/IRX5, reactivated browning programs, and increased thermogenesis Links obesity genetics to PCOS metabolic featuresShows direct manipulation of the FTO risk allele in primary human adipocytes, and highlights its ability to repress thermogenesis in adipocytes, independent of the central nervous system.
DENND1A.V2 [57] Human theca-like cellsOverexpression of DENND1A.V2 enhanced CYP17A1 expression and androgen outputResults mirror PCOS-associated hyperandrogenismThe model functionally reproduces the hyperandrogenic PCOS phenotype; however, the mechanism by which DENND1A.V2 regulates CYP17A1 expression remains unclear. However, only one variant was tested, limiting the scope.
CYP17A1 [119]Mouse ovarian theca cellsOverexpression of CYP17A1 resulted in hyperandrogenism, prolonged estrous cycles, and cystic follicle morphologyFeatures mirror clinical and histologic findings in PCOSOvary-specific overexpression enables temporal control of androgen excess and closely reproduces hyperandrogenic PCOS features. However, systemic metabolic effects were mild, and interspecies differences may limit full translational applicability.
AMH [123,124] Mouse testes and ovariesCRISPR/Cas9 deletion of GATA-binding site in AMH promoter reduced AMH expression in fetal/neonate testes but not in adult ovariesInsights into AMH transcriptional control can help explain dysregulated folliculogenesis.The model demonstrates site-specific control, which facilitates the identification of transcriptional regulatory mechanisms rather than total gene loss. Since the deletion affected fetal/neonatal testes and not adult ovaries, the model cannot fully represent AMH dysregulation throughout life.
AKT2 [164]MiceKnockout mice exhibited growth deficiency, lipoatrophy, IR, hyperglycemia, hyperinsulinemia, and impaired muscle glucose uptakeDemonstrates AKT2’s role in insulin signaling, glucose metabolism, and adipose regulationEnabled dissection of tissue- and sex-specific AKT2 functions, particularly in adipose tissue and pancreatic β-cells. The model also mimicked human type 2 diabetes progression, offering translational insight into β-cell compensation and failure. However, the phenotype was strain-dependent, reducing reproducibility across genetic backgrounds.
LEPR [168] Loss of leptin receptor caused hyperphagia, obesity, hyperglycemia, IR, dyslipidemia, and glucose intoleranceEstablishes leptin signaling as key to metabolic dysfunction seen in PCOSThe models successfully reproduce the hallmark metabolic disturbances seen in obesity and diabetes, and demonstrate high editing efficiency of CRISPR/Cas9, producing fully penetrant phenotypes without the need for multiple breeding generations. Yet, the line could not be maintained as the mice were severely obese and infertile.
LNK (SH2B3) [183]LNK knockout group exhibited a partially restored estrous cycle and an improved glucose metabolismLNK may be a target for PCOS clinical treatmentThe model combines genetic (LNK knockout) and hormonal (DHEA + high-fat diet) approaches, replicating both ovarian and metabolic abnormalities. Limitations include the unclear mechanism of FOXO3 (a transcription factor), and hyperandrogenism was underexplored.
IGFBP7 [187] IGFBP7 is linked to cystic follicular enlargement, reduced granulosa cell layers, immature and atretic follicles, and the disappearance of oocytes. Knockout of the IGFBP7 gene reversed the disruption caused by the administered DHEAIGFBP7 plays a role in the development of PCOS Combines genetic (IGFBP7 knockout) and hormonal (DHEA) models, and uses spleen transcriptomics to uncover immune-endocrine interplay. However, focus was on spleen immune cells, not ovarian tissue.
CYP19A1A [155,156]Zebrafish Knockout of CYP19A1A abolished estrogen production, resulting in complete ovary-to-testis sex reversal and infertilityHighlights the role of estrogen in establishing and maintaining female reproductive architecture and follicle progressionSuccessful in vivo monitoring of gonadal differentiation. However, an off-targeting effect may occur due to the short recognition site of 18 nucleotides.
ADAMTS9 [157] Knockout of ADAMTS9 led to underdeveloped ovaries composed entirely of early-stage oocytes that failed to ovulateDemonstrates ADAMTS9’s role in follicle rupture and oocyte maturationThe ADAMTS9 knockout zebrafish can survive to adulthood, allowing for in vivo study. However, the zebrafish exhibited a strong sex bias towards males, leaving a small number of homozygous knockout females in the offspring of heterozygous crossings.
GSDF [179]Knockout female zebrafish exhibited infertility, oligo-ovulation, hyperandrogenism, IR, and obesityMetabolic and reproductive phenotypes of PCOS were displayedThe model revealed a clear and reproducible phenotype, where all gsdf mutant fish developed as females, but it lacked rescue experiments to confirm specificity of the phenotype.
BMPR1B [149]Goat granulosa cellsKnockout of BMPR1B enhanced SMAD signaling, made cells more sensitive to gonadotropins, reduced cell growth/viability, and removed the usual BMP-4/7 suppression of progesterone productionShows the gene’s importance in regulating granulosa cell growth and hormone productionThe model successfully mimicked the biological phenotype seen in FecB-positive sheep, and used a primary granulosa cell culture, offering a controlled environment to directly assess Smad signaling and steroidogenesis. However, the model does not replicate systemic hormonal interactions, folliculogenesis, or ovulation seen in vivo.
MC4R [170] PigsMC4R-deficient pigs developed marked hyperinsulinemia, IR, hyperorexia, hyperphagia, increased adiposity, and dysregulated lipid metabolism with substantial hepatic fat accumulationHighlights melanocortin pathway’s role in obesity-related metabolic dysfunction relevant to PCOSThe model offers a better physiological and metabolic similarity to humans, but failed to assess long-term disease progression.
IR: insulin resistance; ARID5B: AT-rich interactive domain-containing protein 5B; DHEA: dehydroepiandrosterone.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Bucheeri, S.; Alcibahy, Y.; Bucheeri, Y.; Bucheeri, S.; Alhermi, A.; Butler, A.E. CRISPR as a Tool to Uncover Gene Function in Polycystic Ovary Syndrome: A Literature Review of Experimental Models Targeting Ovarian and Metabolic Genes. Cells 2025, 14, 1769. https://doi.org/10.3390/cells14221769

AMA Style

Bucheeri S, Alcibahy Y, Bucheeri Y, Bucheeri S, Alhermi A, Butler AE. CRISPR as a Tool to Uncover Gene Function in Polycystic Ovary Syndrome: A Literature Review of Experimental Models Targeting Ovarian and Metabolic Genes. Cells. 2025; 14(22):1769. https://doi.org/10.3390/cells14221769

Chicago/Turabian Style

Bucheeri, Shahd, Yasmine Alcibahy, Yara Bucheeri, Sarah Bucheeri, Abrar Alhermi, and Alexandra E. Butler. 2025. "CRISPR as a Tool to Uncover Gene Function in Polycystic Ovary Syndrome: A Literature Review of Experimental Models Targeting Ovarian and Metabolic Genes" Cells 14, no. 22: 1769. https://doi.org/10.3390/cells14221769

APA Style

Bucheeri, S., Alcibahy, Y., Bucheeri, Y., Bucheeri, S., Alhermi, A., & Butler, A. E. (2025). CRISPR as a Tool to Uncover Gene Function in Polycystic Ovary Syndrome: A Literature Review of Experimental Models Targeting Ovarian and Metabolic Genes. Cells, 14(22), 1769. https://doi.org/10.3390/cells14221769

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

Article Metrics

Back to TopTop